bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒08‒11
73 papers selected by
Gavin McStay
Staffordshire University


  1. Neurochem Res. 2019 Aug 05.
      Methamphetamine (METH) is a potent psychomotor stimulant that has a high potential for abuse in humans. In addition, it is neurotoxic, especially in dopaminergic neurons. Long-lasting exposure to METH causes psychosis and increases the risk of Parkinson's disease. Apelin-13 is a novel endogenous ligand which studies have shown that may have a neuroprotective effect. Therefore, we hypothesized that Apelin-13 might adequately prevent METH-induced neurotoxicity via the inhibition of apoptotic, autophagy, and ROS responses. In this study, PC12 cells were exposed to both METH (0.5, 1, 2, 3, 4, 6 mmol/L) and Apelin-13 (0.5, 1.0, 2.0, 4.0, 8.0 μmol/L) in vitro for 24 h to measure determined dose, and then downstream pathways were measured to investigate apoptosis, autophagy, and ROS responses. The results have indicated that Apelin-13 decreased the apoptotic response post-METH exposure in PC12 cells by increasing cell viability, reducing apoptotic rates. In addition, the study has revealed Apelin-13 decreased gene expression of Beclin-1 by Real-Time PCR and LC3-II by western blotting in METH-induced PC12 cells, which demonstrated autophagy is reduced. In addition, this study has shown that Apelin-13 reduces intracellular ROS of METH-induced PC12 cells. These results support Apelin-13 to be investigated as a potential drug for treatment of neurodegenerative diseases. It is suggested that Apelin-13 is beneficial in reducing oxidative stress, which may also play an important role in the regulation of METH-triggered apoptotic response. Hence, these data indicate that Apelin-13 could potentially alleviate METH-induced neurotoxicity via the reduction of oxidative damages, apoptotic, and autophagy cell death.
    Keywords:  Apelin-13; Apoptosis; Autophagy; Methamphetamine; Neurotoxicity; Reactive oxygen species
    DOI:  https://doi.org/10.1007/s11064-019-02847-9
  2. Life Sci. 2019 Aug 05. pii: S0024-3205(19)30662-9. [Epub ahead of print] 116735
      AIMS: The present study was to investigate the protective effects of Zn supplementation in OTA-induced apoptosis of Madin-Darby canine kidney (MDCK) epithelial cells and explore the potential mechanisms. Aiming to provides a new insight into the treatment strategy of OTA-induced nephrotoxicity by nutritional regulation.MAIN METHODS: Initially, through MTT and LDH assay revealed that Zn supplementation significantly suppressed OTA-induced cytotoxicity in MDCK cells. Then, the production of reactive oxygen species (ROS) was detected by using a DCFH-DA assay. Annexin V-FITC/PI, Hoechst 33258 staining and Flow cytometry were used to detect the apoptosis. The expressions of apoptosis-related molecules were determined by RT-PCR, Western blotting. Interestingly, OTA treatment slightly increased the levels of Metallothionein-1 (MT-1) and Metallothionein-2 (MT-2) by using RT-PCR, Western blotting assay; while Zn supplementation further improved the increase of MT-1 and MT-2 induced by OTA. However, the inhibitive effects of Zn supplementation were significantly blocked after double knockdown of MT-1 and MT-2 by using Small Interfering RNA (siRNA) Transfection method.
    KEY FINDINGS: Our study provides supportive data for the potential roles of Zn in reducing OTA-induced oxidative stress and apoptosis in MDCK cells.
    SIGNIFICANCE: Zn is one of the key structural components of many proteins, which plays an important role in several physiological processes such as cell survival and apoptosis. This metal is expected to contribute to the conservative and adjuvant treatment of kidney disease and should therefore be investigated further.
    Keywords:  Apoptosis; MDCK cells; Metallothioneins; Ochratoxin A; Zinc
    DOI:  https://doi.org/10.1016/j.lfs.2019.116735
  3. Biomolecules. 2019 Aug 05. pii: E346. [Epub ahead of print]9(8):
      Cyclophosphamide (CP) is a widely used chemotherapeutic agent; however, its clinical application is limited because of its multi-organ toxicity. Galangin (Gal) is a bioactive flavonoid with promising biological activities. This study investigated the hepatoprotective effect of Gal in CP-induced rats. Rats received Gal (15, 30 and 60 mg/kg/day) for 15 days followed by a single dose of CP at day 16. Cyclophosphamide triggered liver injury characterized by elevated serum transaminases, alkaline phosphatase (ALP) and lactate dehydrogenase (LDH), and histopathological manifestations. Increased hepatic reactive oxygen species, malondialdehyde, nitric oxide, and oxidative DNA damage along with declined glutathione and antioxidant enzymes were demonstrated in CP-administered rats. CP provoked hepatic nuclear factor-kappaB (NF-κB) phosphorylation and increased mRNA abundance of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), and tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) both expression and serum levels. Gal prevented CP-induced liver injury, boosted antioxidants and suppressed oxidative stress, DNA damage, NF-κB phosphorylation and pro-inflammatory mediators. Gal diminished Bax and caspase-3, and increased B-cell lymphoma-2 (Bcl-2) in liver of CP-administered rats. In addition, Gal increased peroxisome proliferator-activated receptor gamma (PPARγ) expression and activated hepatic nuclear factor erythroid 2-related factor 2 (Nrf2) signaling showed by the increase in Nrf2, NAD(P)H: quinone acceptor oxidoreductase-1 (NQO-1) and heme oxygenase 1 (HO-1) in CP-administered rats. These findings suggest that Gal prevents CP hepatotoxicity through activation of Nrf2/HO-1 signaling and attenuation of oxidative damage, inflammation and cell death. Therefore, Gal might represent a promising adjuvant therapy to prevent hepatotoxicity in patients on CP treatment.
    Keywords:  cyclophosphamide; galangin; hepatotoxicity; inflammation; nuclear factor erythroid 2-related factor 2; reactive oxygen species
    DOI:  https://doi.org/10.3390/biom9080346
  4. Med Microbiol Immunol. 2019 Aug 03.
      Pseudomonas aeruginosa is an opportunistic pathogen that utilizes the quorum-sensing (QS) process to regulate the production of different virulence factors and biofilm. N-3-oxo-dodecanoyl-L-homoserine lactone (C12) is a key QS molecule of P. aeruginosa which interacts with the mammalian immune cells and modulates their function. Here, we investigated the molecular mechanism of C12-induced apoptosis in neutrophils. Our data show that C12 causes apoptosis in neutrophils through an elevation in cytosolic and mitochondrial Ca2+ levels. Besides, C12 induces phosphatidylserine (PS) exposure, mitochondrial membrane potential (MMP) depolarization, mitochondrial permeability transition pore (MPTP) formation and mitochondrial reactive oxygen species (mROS) generation. C12-induced rise in intracellular Ca2+ level is majorly contributed by endoplasmic reticulum store through the activation of inositol 1, 4, 5-triphosphate receptor. Intracellular calcium chelation inhibited C12-induced mitochondrial dysfunction and apoptosis. Further, inhibition of mitochondrial Ca2+ uniporter by ruthenium red or Ru360 abrogated C12-induced mitochondrial Ca2+ uptake, MMP loss, MPTP opening, mROS production, and PS exposure. These mechanistic insights are expected to provide a better understanding of the role of C12 in P. aeruginosa pathogenesis.
    Keywords:  Apoptosis; Calcium signaling; Mitochondria; Neutrophil; ROS; mCU
    DOI:  https://doi.org/10.1007/s00430-019-00631-8
  5. Eur Rev Med Pharmacol Sci. 2019 Aug;pii: 18660. [Epub ahead of print]23(3 Suppl): 294-303
      OBJECTIVE: To clarify the role of aldose reductase inhibitor (ARI) in the high glucose-induced cardiomyocyte apoptosis and its mechanism.MATERIALS AND METHODS: In this study, H9c2 cardiomyocytes were employed as objects, high-glucose medium as stimulus, and ARI Epalrestat as a therapeutic drug. The cell apoptosis and activity changes of nitric oxide synthase (NOS), NO, and reactive oxygen species (ROS) were evaluated via Hoechst staining, enzyme-linked immunosorbent assay (ELISA), polymerase chain reaction (PCR), and Western blotting. In addition, the mitochondrial membrane potential was measured via fluorescence counting.
    RESULTS: Epalrestat inhibited the activity of AR to improve high glucose-induced oxidative stress in cardiomyocytes, weaken ROS activity, relieve the inhibition on NO activity, alleviate mitochondrial membrane potential damage, reduce the level of high glucose-induced cardiomyocyte apoptosis, and suppress the expression and activity of Caspase-3, thereby preventing high glucose-induced cardiomyocyte apoptosis.
    CONCLUSIONS: ARI protects against high glucose-induced cardiomyocyte apoptosis.
    DOI:  https://doi.org/10.26355/eurrev_201908_18660
  6. Biochim Biophys Acta Mol Basis Dis. 2019 Jul 31. pii: S0925-4439(19)30240-6. [Epub ahead of print]
      Dermal fibroblasts are mesenchymal cells found between the skin epidermis and subcutaneous tissue that play a pivotal role in cutaneous wound healing by synthesizing fibronectin (a component of the extracellular matrix), secreting angiogenesis factors, and generating strong contractile forces. In wound healing, low concentrations of reactive oxygen species (ROS) are essential in combating invading microorganisms and in cell-survival signaling. However, excessive ROS production impairs fibroblasts. Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) is a key enzyme that regulates the mitochondrial redox balance and reduces oxidative stress-induced cell injury through the generation of NADPH. In the present study, the downregulation of IDH2 expression resulted in an increase in cell apoptosis in mouse skin through ROS-dependent ATM-mediated p53 signaling. IDH2 deficiency also delayed cutaneous wound healing in mice and impaired dermal fibroblast function. Furthermore, pretreatment with the mitochondria-targeted antioxidant mito-TEMPO alleviated the apoptosis induced by IDH2 deficiency both in vitro and in vivo. Together, our findings highlight the role of IDH2 in cutaneous wound healing in association with mitochondrial ROS.
    Keywords:  Apoptosis; IDH2; Mito-TEMPO; Mitochondria; Wound healing
    DOI:  https://doi.org/10.1016/j.bbadis.2019.07.017
  7. Cell Death Dis. 2019 Aug 08. 10(8): 600
      Oxaliplatin-based chemotherapy is recommended as the first-line therapeutic regimen for metastatic colorectal cancer. However, long-term and repeated oxaliplatin therapy leads to drug resistance and severe adverse events, which hamper its clinical application. Thus, chemosensitizers are urgently required for overcoming oxaliplatin resistance and toxicity. Here, the anticancer effects of oxaliplatin combined with piperlongumine (PL), a molecule promoting reactive oxygen species (ROS) generation, in colorectal cancer, were assessed. We demonstrated that oxaliplatin elevated cellular ROS amounts and showed synergistic anticancer effects with PL in colorectal cancer cells. These anticancer effects were mediated by mitochondrial dysfunction and endoplasmic reticulum (ER) stress apoptotic-associated networks. Meanwhile, blockage of ROS production prevented apoptosis and fully reversed mitochondrial dysfunction and ER stress associated with the oxaliplatin/PL combination. Moreover, xenograft assays in mouse models highly corroborated in vitro data. In conclusion, this study provides a novel combination therapy for colorectal cancer, and reveals that manipulating ROS production might constitute an effective tool for developing novel treatments in colorectal cancer.
    DOI:  https://doi.org/10.1038/s41419-019-1824-6
  8. Reprod Biol. 2019 Aug 03. pii: S1642-431X(19)30172-X. [Epub ahead of print]
      Granulosa Cells (GCs) are sensitive to excessive production of reactive oxygen species (ROS). Quercetin (QUR) is a free radical scavenger which can alleviate oxidative stress through nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/antioxidant response element (ARE) pathway and thioredoxin (Trx) system. We aimed to explore the probable protective role of QUR on cultured human GCs treated with hydrogen peroxide (H2O2) as an inducer of oxidative stress. MTT assay was applied for evaluating the cell cytotoxicity of QUR and H2O2. The rate of apoptotic cells and intracellular ROS generation were determined by Annexin V-FITC/PI staining and 2'-7'-dichlorodihydrofluorescein diacetate fluorescent probes (DCFH-DA), respectively. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis and western blot analysis were used to evaluate the gene and protein expression of Nrf2 and kelch-like ech-associated protein 1 (Keap1)1. The Nrf2 and Trx activities were measured by Enzyme-linked Immunosorbent Assay (ELISA). The results indicated that QUR pretreatment can decrease ROS production and apoptosis induced by H2O2. In addition, QUR increased Nrf2 gene and protein expression, as well as its nuclear translocation. Moreover, in QUR-treated group, a lower level of Keap1 protein was observed, which was not reported as significant. The results also indicated a significant correlation between the expression of Nrf2 and Keap1 in QUR-treated group. Further, QUR protected GCs from oxidative stress by increasing Trx gene expression and activity. This study suggests that QUR as a supplementary factor may protect GCs from oxidative stress in diseases related to this condition.
    Keywords:  Granulosa cells; Nuclear factor (erythroid-derived 2)-like 2; Quercetin; Reactive oxygen species; Thioredoxin
    DOI:  https://doi.org/10.1016/j.repbio.2019.07.002
  9. Acta Pharm Sin B. 2019 Jul;9(4): 690-701
      Oxidative stress and cardiomyocyte apoptosis are involved in the pathogenesis of doxorubicin (DOX)-induced cardiotoxicity. Matrine is well-known for its powerful anti-oxidant and anti-apoptotic capacities. Our present study aimed to investigate the effect of matrine on DOX-induced cardiotoxicity and try to unearth the underlying mechanisms. Mice were exposed with DOX to generate DOX-induced cardiotoxicity or normal saline as control. H9C2 cells were used to verify the effect of matrine in vitro. DOX injection triggered increased generation of reactive oxygen species (ROS) and excessive cardiomyocyte apoptosis, which were significantly mitigated by matrine. Mechanistically, we found that matrine ameliorated DOX-induced uncoupling protein 2 (UCP2) downregulation, and UCP2 inhibition by genipin could blunt the protective effect of matrine on DOX-induced oxidative stress and cardiomyocyte apoptosis. Besides, 5'-AMP-activated protein kinase α2 (Ampkα2) deficiency inhibited matrine-mediated UCP2 preservation and abolished the beneficial effect of matrine in mice. Besides, we observed that matrine incubation alleviated DOX-induced H9C2 cells apoptosis and oxidative stress level via activating AMPKα/UCP2, which were blunted by either AMPKα or UCP2 inhibition with genetic or pharmacological methods. Matrine attenuated oxidative stress and cardiomyocyte apoptosis in DOX-induced cardiotoxicity via maintaining AMPKα/UCP2 pathway, and it might be a promising therapeutic agent for the treatment of DOX-induced cardiotoxicity.
    Keywords:  4-HNE, 4-hydroxynonenal; ACC, acetyl-CoA carboxylase; AMPKα; AMPKα, 5′-AMP-activated protein kinase α; ANOVA, analysis of variance; Apoptosis; BAX, BCL-2-associated X protein; BCA, bicinchoninic acid; BCL-2, B-cell lymphoma 2; C-caspase 3, cleaved-caspase3; CCK-8, cell counting kit 8; CK-MB, creatine kinase isoenzymes; DCFH-DA, 2′,7′-dichlorodihydrofluorescein diacetate; DHE, dihydroethidium; DMEM, Dulbecco׳s modified Eagle׳s medium; DOX, doxorubicin; FBS, fetal bovine serum; FS, fractional shortening; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; HW, heart weight; LDH, lactate dehydrogenase; MDA, malondialdehyde; Matrine; Oxidative stress; PPARs, peroxisomal proliferators-activated receptors; ROS, reactive oxygen species; SOD2, superoxide dismutase 2; T-caspase3, total-caspase3; TL, tibia length; TUNEL, TdT-mediated dUTP nick end-labelling; Top2, topoisomerase-II; UCP2; UCP2, uncoupling protein 2; cTnT, cardiac isoform of Tropnin T
    DOI:  https://doi.org/10.1016/j.apsb.2019.03.003
  10. Poult Sci. 2019 Aug 09. pii: pez465. [Epub ahead of print]
      Avian infectious bronchitis virus (IBV), a coronavirus, causes infectious bronchitis leading to enormous economic loss in the poultry industry worldwide. Hypericin (HY) is an excellent compound that has been investigated in antiviral, antineoplastic, and antidepressant. To investigate the inhibition effect of HY on IBV infection in chicken embryo kidney (CEK) cells, 3 different experimental designs: pre-treatment of cells prior to IBV infection, direct treatment of IBV-infected cells, and pre-treatment of IBV prior to cell infection were used. Quantitative real-time PCR (qRT-PCR), immunofluorescence assay (IFA), flow cytometry, and fluorescence microscopy were performed and virus titer was determined by TCID50. The results revealed that HY had a good anti-IBV effect when HY directly treated the IBV-infected cells, and virus infectivity decreased in a dose-dependent manner. Furthermore, HY inhibited IBV-induced apoptosis in CEK cells, and significantly reduced the mRNA expression levels of Fas, FasL, JNK, Bax, Caspase 3, and Caspase 8, and significantly increased Bcl-2 mRNA expression level in CEK cells. In addition, HY treatment could decrease IBV-induced reactive oxygen species (ROS) generation in CEK cells. These results suggested that HY showed potential antiviral activities against IBV infection involving the inhibition of apoptosis and ROS generation in CEK cells.
    Keywords:  apoptosis; chicken embryo kidney cells; hypericin; infectious bronchitis virus; reactive oxygen species
    DOI:  https://doi.org/10.3382/ps/pez465
  11. Radiat Res. 2019 Aug 07.
      Photodynamic therapy (PDT) uses a combination of photosensitizers with visible light to generate reactive species and selectively kill tumor or unwanted tissue. Znln2S4 nanoparticles are widely implemented in photovoltaic device materials and photolysis water catalysts owing to their unique photoelectric properties. Whether Znln2S4 itself can be used as an effective dye in PDT is still unknown. To determine the effects and potential mechanism of Znln2S4-PDT on HepG2 cell apoptosis, electron microscopic analysis was performed to monitor the apoptotic morphology of HepG2 cells upon exposure to Znln2S4-PDT. Flow cytometry was performed to measure the apoptosis rate and intracellular ROS production. Western blot and ELISA were performed to reveal the expression changes in Bax, caspase-3 and caspase-9. Data from this work suggested that cells exhibited the typical apoptotic morphology in response to Znln2S4-PDT, with high apoptotic rate. The intracellular ROS production after Znln2S4-PDT occurred in a dose-dependent manner. Moreover, Znln2S4-PDT augmented the expression levels of pro-apoptosis factors, especially, Bax, caspase-3 and caspase-9. Taken together, our novel findings, Znln2S4-PDT elicited HepG2 cell apoptosis, suggesting Znln2S4 as a promising photosensitizer candidate for cancer therapy.
    DOI:  https://doi.org/10.1667/RR15389.1
  12. Biomed Pharmacother. 2019 Sep;pii: S0753-3322(19)31812-8. [Epub ahead of print]117 109143
      Araloside C (AsC) has potential cardioprotective properties. However, the underlying mechanism of AsC-mediated cardioprotection, especially the role of mitochondrial function, remains largely unknown. Here, we used H9c2 cardiomyocytes to study the cardioprotective mechanisms of AsC through H2O2-induced oxidative stress. Cell viability, lactate dehydrogenase release, mitochondrial functions and bioenergetics were evaluated. Western blot analysis was used to measure the protein expression levels of apoptosis and the phosphorylation of AMP-activated protein kinase (AMPK). Results revealed that AsC increased cell viability, improved mitochondrial membrane potential disruption, decreased mitochondrial reactive oxygen species level, elevated cellular ATP levels and alleviated impaired mitochondrial respiration in H2O2-induced H9c2 cardiomyoblasts injury. Furthermore, AsC modulated apoptosis-associated protein expression and AMPK pathway in H9c2 cells under oxidative stress. In conclusion, AsC potentially protects H9c2 cardiomyoblasts against oxidative stress by regulating mitochondrial function and AMPK activation. AsC may be an effective therapeutic agent for the prevention of oxidative stress in cardiac injury.
    Keywords:  AMPK; Araloside C; Cardioprotection; Mitochondrial function
    DOI:  https://doi.org/10.1016/j.biopha.2019.109143
  13. Neurotoxicology. 2019 Aug 02. pii: S0161-813X(19)30076-2. [Epub ahead of print]74 221-229
      Triclosan (TCS) has been widely used as a disinfectant and antiseptic in multiple consumer and healthcare products due to its clinical effectiveness against various bacteria, fungi and protozoa. Recently, several studies have reported the adverse effects of TCS on various nerve cells, arousing concerns about its potential neurotoxicity. The present study aimed to investigate the neurotoxicity of TCS in rat pheochromocytoma PC12 cells. After differentiation, the stabilized PC12 cells were treated with 1, 10, 50 μM TCS for 12 h. At the end of the treatment, the generation of reactive oxygen species (ROS), protein expression of apoptotic-related genes, AMPK-AKT/mTOR, as well as p38 in PC12 cells were determined. The concentrations were chosen based on the results of cell viability and lactic dehydrogenase (LDH) assays in response to TCS treatment (ranging from 0.001 to 100 μM) for varied time periods. The results showed that TCS is cytotoxic to PC12 cells, causing decreased cell viability accompanied by increased LDH release. TCS treatment at 10 and 50 μM for 12 h increased the mRNA and protein expression of the pro-apoptotic gene Bax, while Bcl-2 levels remained unchanged. Moreover, an increase in the generation of reactive oxygen species (ROS) was found in TCS-treated PC12 cells at the concentrations of 1 and 10 μM. Pretreatment with 100 μM N-acetyl cysteine (NAC- ROS scavenger) for 1 h normalized the ROS generations in TCS-treated PC12 cells. Additionally, the suppression of the phosphorylation of Akt and mTOR was observed in TCS-treated PC12 cells at 10 and 50 μM for 12 h, concomitant with the activation of p38 MAPK pathway at 50 μM TCS. However, there were no effects of TCS on the phosphorylation of AMPK in these cells. Taken together, these results suggest that TCS may cause adverse effects and oxidative stress in PC12 cells accompanied by inhibition of Akt/mTOR and activation of p38.
    Keywords:  AKT/mTOR pathway; Apoptosis; PC12; Triclosan; p38
    DOI:  https://doi.org/10.1016/j.neuro.2019.07.008
  14. Toxicology. 2019 Aug 02. pii: S0300-483X(19)30221-5. [Epub ahead of print] 152267
      Paraquat has relatively strong detrimental effects on humans and animals and can cause acute lung injury with high mortality. Ghrelin is a brain-gut peptide which plays important roles in regulating various physiological processes. This study investigated whether ghrelin could inhibit paraquat-induced lung injuries and attempted to elucidate the possible molecular mechanisms. A549 cells were preincubated with different concentrations of ghrelin and then treated with 200 μM of PQ for 24 h. Then cell survival, apoptosis, cellular oxidative stress and lipid peroxidation of A549 cells were detected after different treatments. Subsequently, we analyzed the mitochondrial membrane potential (ΔΨm) and measured caspase-3 activation in A549 cells. In addition, we investigated the activation of the MAPKs pathway and the function of p38-MAPK within mitochondrial apoptosis. Our study indicated that ghrelin administration improved cell viability and reduced apoptosis of PQ-treated A549 cells dose-dependently. Ghrelin treatment reduced the elevation of ROS and MDA, while improved GSH content in A549 cells after paraquat exposure. Moreover, we found that ghrelin dose-dependently increased ΔΨm and decreased caspase-3 activity. The phosphorylated p38 MAPK and JNK levels elevated following PQ exposure, while the phosphorylation of p38 MAPK decreased following ghrelin pretreatment. p38 MAPK siRNA or SB203580 pretreatment ameliorated PQ-caused cell injury and apoptosis related signals, however, the intracellular ROS production was not affected. N-acetylcysteine (NAC), a classic antioxidant pretreatment decreased the phosphorylated p38 MAPK level and intracellular ROS production, alleviated cell injury, and inhibited apoptosis. The results showed that p38-MAPK pathway plays an important role in PQ-caused alveolar epithelial cell insult, and ghrelin might attenuate PQ-induced cell injury by inhibiting ROS-induced p38-MAPK modulated mitochondrial apoptotic pathway.
    Keywords:  Acute lung injury; Apoptosis; Ghrelin; Paraquat; p38-MAPK
    DOI:  https://doi.org/10.1016/j.tox.2019.152267
  15. Endocr Metab Immune Disord Drug Targets. 2019 Aug 06.
      BACKGROUND: Nowadays the potential therapeutic role of various bioflavonoids including Curcumin, Luteolin and Resveratrol has currently been well-documented in a vast range of fatal complications including synaptic failure and cancers. These bioflavonoids are widely being implemented in the treatment of various cancers as these possess anti-cancerous, anti-oxidant and anti-inflammatory properties. Moreover, these are also used as a better alternative to conventional therapies since; these are non-toxic to cells and having no or least side effects. Notably, the pertinent therapeutic role of Rutin in cervical cancer is still unsettled however its anti-cancerous role has already been reported in other cancers including prostate and colon cancer. Rutin (Vitamin P or Rutoside) is a polyphenolics flavonoid exhibiting multi-beneficial roles against several carcinomas.OBJECTIVE: Despite the evidence for its several biological activities, the anticancer effects of Rutin on human cervical cancer (C33A) cells remain to be explored. In this study, the anticancer potential of Rutin was investigated by employing the key biomarkers such as nuclear condensation reactive oxygen species (ROS), apoptosis, and changes in mitochondrial membrane potential (MMP).
    RESULTS: Our findings showed that Rutin treatment reduced the cell viability, induced significant increase in ROS production and nuclear condensation in dose dependent manner. Moreover, Rutin provoked apoptosis by inducing decrease in MMP and activation of caspase-3. Cell cycle analysis further confirmed the efficacy of Rutin by showing cell cycle arrest at G0/G1 phase.
    CONCLUSION: Thus, our study is envisaged to open up interests for elucidating Rutin as an anti-cancerous agent against cervical cancer.
    Keywords:  C33A; Cervical cancer; ROS; apoptosis; cell cycle; rutin
    DOI:  https://doi.org/10.2174/1871530319666190806122257
  16. Int J Mol Sci. 2019 Aug 06. pii: E3836. [Epub ahead of print]20(15):
      5-Lipoxygenase (5-LO) has been reported to be highly expressed in brain tumors and to promote glioma cell proliferation. Therefore, we investigated the anticancer activity of the novel 5-LO inhibitor derivative 3-tridecyl-4,5-dimethoxybenzene-1,2-diol hydroquinone (EA-100C red) on glioblastoma (GBM) cell growth. Cell viability was evaluated by MTT assay. The effects of the compound on apoptosis, oxidative stress and autophagy were assessed by flow cytometry (FACS). The mode of action was confirmed by Taqman apoptosis array, Real Time qPCR, confocal microscopy analysis and the western blotting technique. Our results showed that EA-100C Red had a higher anti-proliferative effect on LN229 as compared to U87MG cells. The compound induced a significant increase of apoptosis and autophagy and up-regulated pro-apoptotic genes (Bcl3, BNIP3L, and NFKBIA) in both GBM cell lines. In this light, we studied the effects of EA-100C red on the expression of CHOP and XBP1, that are implicated in ER-stress-mediated cell death. In summary, our findings revealed that EA-100C red induced ER stress-mediated apoptosis associated to autophagy in GBM cells through CHOP and Beclin1 up-regulation and activation of caspases 3, 9, JNK and NF-kappaB pathway. On these bases, EA-100C red could represent a promising compound for anti-cancer treatment.
    Keywords:  ER stress; anticancer drug; apoptosis; autophagy; natural compound; reactive oxygen species (ROS)
    DOI:  https://doi.org/10.3390/ijms20153836
  17. Cell Biol Int. 2019 Aug 08.
      Cardamonin (CD), a naturally occurring chalcone isolated from large black cardamom, was previously reported to suppress the proliferation of breast cancer cells. However, its precise molecular antitumor mechanisms have not been well elucidated. In this study, we found that CD markedly inhibited the proliferation of MDA-MB 231 and MCF-7 breast cancer cells through the induction of G2/M arrest and apoptosis. Reactive oxygen species (ROS) plays a pivotal role in the inhibition of CD-induced cell proliferation. Treatment with N-acetyl-cysteine (NAC), an ROS scavenger, blocked CD-induced G2/M arrest and apoptosis in this study. Quenching of ROS by overexpression of catalase also blocked CD-induced cell cycle arrest and apoptosis. We showed that CD enhanced the expression and nuclear translocation of Forkhead box O3 (FOXO3a) via upstream c-Jun N-terminal kinase, inducing the expression of FOXO3a and its target genes, including p21, p27, and Bim. This process led to the reduction of cyclin D1 and enhancement of activated caspase-3 expression. The addition of NAC markedly reversed these effects, knockdown of FOXO3a using small interfering RNA also decreased CD-induced G2/M arrest and apoptosis. In vivo, CD efficiently suppressed the growth of MDA-MB 231 breast cancer xenograft tumors. Taken together, our data provide a molecular mechanistic rationale for CD-induced cell cycle arrest and apoptosis in breast cancer cells. This article is protected by copyright. All rights reserved.
    Keywords:  Breast cancer; Cardamonin; FOXO3a; JNK; ROS
    DOI:  https://doi.org/10.1002/cbin.11217
  18. Life Sci. 2019 Aug 05. pii: S0024-3205(19)30659-9. [Epub ahead of print] 116732
      AIMS: Linderane, an important bioactive compound in Linderae, improved glucose and lipid metabolism in ob/ob mice. However, the effect of linderane on streptozotocin (STZ)-induced oxidative damage in INS-1 cells remains unclear.MAIN METHODS: INS-1 cells were pre-treated with different doses of linderane for 2 h and then treated with 3 mM STZ for 12 h. Cell viability was determined by MTT assay. Cell apoptosis was detected using an Annexin V-FITC Apoptosis Detection Kit. The level of intracellular ROS was determined using dichlorofluorescein-diacetate (DCFH-DA). The activities of insulin secretion, SOD, catalase (CAT) and GPx were measured using ELISA kits. The expression levels of bax, bcl-2, p38, p-p38, nuclear Nrf2 and HO-1 were measured using western blot.
    KEY FINDINGS: The results showed that STZ-caused inhibitory effects on cell viability and insulin secretion were mitigated by linderane. Furthermore, linderane inhibited apoptosis and oxidative stress in STZ-induced INS-1 cells. Finally, linderane suppressed the activation of p38 MAPK pathway, as well as enhanced the activation of Nrf2 pathway in STZ-induced INS-1 cells. Activation of p38 MAPK pathway or inhibition of Nrf2 significantly reversed the protective effects of linderane against STZ-induced ROS production and cell apoptosis.
    SIGNIFICANCE: The protective effects of linderane on STZ-induced INS-1 cells might be attributed to the inhibition of p38 MAPK and activation of Nrf2 pathway.
    Keywords:  Diabetes mellitus (DM); Linderane; Nrf2 signaling pathway; Oxidative stress; Pancreatic islet β cells; p38 MAPK signaling pathway
    DOI:  https://doi.org/10.1016/j.lfs.2019.116732
  19. Biomed Pharmacother. 2019 Aug 02. pii: S0753-3322(18)37102-6. [Epub ahead of print]118 108589
      (20R)-Dammarane-3β, 12β, 20, 25-tetrol (25-OH-PPD) is a ginsenoside isolated from Panax ginseng (C. A. Meyer). Previous research shows that the compound exhibits anti-cancer activities on many human cancer cell lines. In an attempt to enhance 25-OH-PPD activity, some derivatives were synthesized. Through screening of the derivative compounds for anti-cancer activity against gastric carcinoma cells, 12β-O-(L-Chloracetyl)-dammar-20(22)-ene-3β, 25-diol (4-XL-PPD) was selected as a strong anti-cancer agent. In this study, the anti-cancer mechanisms of 4-XL-PPD were investigated. The results showed that compound 4-XL-PPD resulted in a concentration-dependent inhibition of cells viability in gastric cancer cells, without affecting the viability of normal cell (human gastric epithelial cell line-GES-1). In BGC-803 cancer cells, 4-XL-PPD triggered apoptosis, and stimulated reactive oxygen species production. Apoptosis can be attenuated by the reactive oxygen species scavenger N-acetylcysteine. Meantime, 4-XL-PPD effectively suppressed the migratory and invasive capabilities of BGC-803 cancer cell and inhibited the expression levels of proteins associated with migratory and invasive capabilities (MMP-2, MMP-9, E-cadherin and CD34). All the results suggest that 4-XL-PPD exhibited remarkable anticancer activity base on inducing apoptosis via generating reactive oxygen species and inhibiting migratory and invasive, which support development of 4-XL-PPD as a potential agent for cancer therapy.
    Keywords:  25-OH-PPD; 4-XL-PPD; Apoptosis; Migratory and invasive; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.biopha.2019.01.050
  20. Biomed Pharmacother. 2019 Sep;pii: S0753-3322(19)32301-7. [Epub ahead of print]117 109182
      Bisphenol A (BPA), a widely used industrial compound worldwide, was recently classified as an environmental toxicant. The intestines and liver are responsible for detoxification in humans and animals, and functional damage to these organs adversely affects the health of the body. However, the effect of BPA on intestinal and liver function remains unclear. In this study, we investigated the effects of dietary BPA uptake on oxidative stress, inflammatory response, apoptosis and mitochondrial function in the colons and livers of mice. Dietary BPA uptake significantly reduced the body weights of mice as well as their colon and liver weights. Dietary BPA uptake increased the levels of oxidative stress indicators such as reactive oxygen species, reactive nitrogen species, malondialdehyde and hydrogen peroxide in mouse serum, colon and liver tissues. Antioxidant indicators, such as superoxide dismutase, glutathione peroxidase, catalase and total antioxidant capacity, as well as proinflammatory cytokines (interleukin-1β, interleukin-6, interleukin-8 and tumor necrosis factor-α) were also significantly reduced in the serum, colon, and liver tissues in the BPA group. Moreover, mitochondria-encoded genes and mitochondrial copy numbers were significantly reduced in the colon and liver tissues of the BPA mice. Dietary BPA uptake also increased gene abundance and enzyme activity of caspase-3, -8, -9 and -10. Our study found that dietary BPA induced oxidative stress, inflammatory response, apoptosis and mitochondrial dysfunction in mouse colons and livers.
    Keywords:  Apoptosis; Bisphenol A; Inflammatory response; Mitochondrial function; Oxidative stress
    DOI:  https://doi.org/10.1016/j.biopha.2019.109182
  21. Front Oncol. 2019 ;9 640
      Background: Cisplatin, a powerful antitumor agent, causes formation of DNA adducts, and activation of apoptotic pathways. Presently, cisplatin resistance develops in up to 70% of patients but the underlying molecular mechanism(s) are unclear and there are no markers to determine which patients will become resistant. Mitochondria play a significant role not only in energy metabolism but also retrograde signaling (mitochondria to nucleus) that modulates inflammation, complement, and apoptosis pathways. Maternally inherited mitochondrial (mt) DNA can be classified into haplogroups representing different ethnic populations that have diverse susceptibilities to diseases and medications. Methods: Transmitochondrial cybrids, where all cell lines possess identical nuclear genomes but either the H (Southern European) or J (Northern European) mtDNA haplogroups, were treated with cisplatin and analyzed for differential responses related to viability, oxidative stress, and expression levels of genes associated with cancer, cisplatin-induced nephrotoxicity and resistance, apoptosis and signaling pathways. Results: The cisplatin-treated-J cybrids showed greater loss of cell viability along with lower levels of reactive oxygen species and mitochondrial membrane potential compared to cisplatin-treated-H cybrids. After cisplatin treatment, J cybrids showed increased gene expression of BAX, CASP3, and CYP51A, but lower levels of SFRP1 compared to untreated-J cybrids. The cisplatin-treated-H cybrids had elevated expression of CDKN1A/P21, which has a role in cisplatin toxicity, compared to untreated-H cybrids. The cisplatin-treated H had higher transcription levels of ABCC1, DHRS2/HEP27, and EFEMP1 compared to cisplatin-treated-J cybrids. Conclusions: Cybrid cell lines that contain identical nuclei but either H mtDNA mitochondria or J mtDNA mitochondria respond differently to cisplatin treatments suggesting involvement of the retrograde signaling (from mitochondria to nucleus) in the drug-induced cell death. Varying toxicities and transcription levels of the H vs. J cybrids after cisplatin treatment support the hypothesis that mtDNA variants play a role in the expression of genes affecting resistance and side effects of cisplatin.
    Keywords:  cisplatin; cybrids; drug resistance; mitochondria; mtDNA haplogroups
    DOI:  https://doi.org/10.3389/fonc.2019.00640
  22. Acta Pharm Sin B. 2019 Jul;9(4): 782-793
      The clinical application of doxorubicin (DOX) in cancer chemotherapy is limited by its life-threatening cardiotoxic effects. Chrysophanol (CHR), an anthraquinone compound isolated from the rhizome of Rheum palmatum L., is considered to play a broad role in a variety of biological processes. However, the effects of CHR׳s cardioprotection in DOX-induced cardiomyopathy is poorly understood. In this study, we found that the cardiac apoptosis, mitochondrial injury and cellular PARylation levels were significantly increased in H9C2 cells treated by Dox, while these effects were suppressed by CHR. Similar results were observed when PARP1 activity was suppressed by its inhibitors 3-aminobenzamide (3AB) and ABT888. Ectopic expression of PARP1 effectively blocked this CHR׳s cardioprotection against DOX-induced cardiomyocyte injury in H9C2 cells. Furthermore, pre-administration with both CHR and 3AB relieved DOX-induced cardiac apoptosis, mitochondrial impairment and heart dysfunction in Sprague-Dawley rat model. These results revealed that CHR protects against DOX-induced cardiotoxicity by suppressing cellular PARylation and provided critical evidence that PARylation may be a novel target for DOX-induced cardiomyopathy.
    Keywords:  3AB, 3-aminobenzamide; ADR, adriamycin; ANOVA, one-way analysis of variance; Apoptosis; CHR, chrysophanol; CMC-Na, sodium carboxymethyl; CO, cardiac output; Cardiotoxicity; Chrysophanol; Cyt c, Cytochrome c; DOX, doxorubicin; Doxorubicin; EF, ejection fraction; FBS, fetal bovine serum; FS, fractional shortening; HE, hematoxylin-eosin; HR, heart rate; IVSd, end-diastolic interventricular septum; IVSs, end-systolic interventricular septum; LV, end-systolic volume; LVEDV, LV end-diastolic volume; LVIDd, LV end-diastolic internal diameter; LVIDs, LV end-systolic internal diameter; LVPWd, LV end-diastolic posterior wall thickness; LVPWs, LV end-systolic posterior wall thickness; Mitochondria; NS, normal saline; PAR, polymers of ADP-ribose; PARP1, poly(ADP-ribose) polymerase 1; PARylated, poly(ADP-ribosyl)ated; PARylation; PARylation, poly(ADP-ribosyl)ation; PBS, phosphate-buffered saline; RCR, respiratory control ratio; ROS, reactive oxygen species; Rh123, rhodamine 123; SD, Sprague–Dawley; TUNEL, TdT-mediated dUTP nick end labeling; VDAC1, voltage dependent anion channel 1
    DOI:  https://doi.org/10.1016/j.apsb.2018.10.008
  23. Artif Cells Nanomed Biotechnol. 2019 Dec;47(1): 3297-3305
      Siberian ginseng, perennial herb belongs to Araliaceae family used in traditional medicines to treat hypertension, thrombus, inflammation and cancer. In the present study, we biosynthesized goldnanoparticles using Siberian ginseng aqeous extract in a cost effective manner. The synthesized Siberian ginseng gold nanoparticle (SG-GNPs) were characterized using UV-Vis spec, HR-TEM, XRD, FTIR, SAED analysis. UV-Vis spectroscopic analysis showed a surface Plasmon resonance peak at 538 nm which does not reduce till 30 days of incubation. The results of HR-TEM, XRD and SAED confirm the spherical shape, crystalline nature and the size of the synthesized gold nanoparticles. The FTIR results prove that the biological components present in the Siberian ginseng had reduced the gold ions to synthesis gold nanoparticles. After characterization, the efficacy of SG-GNPS against the melanoma, a deadliest skin carcinoma, was assessed in vitro using B16 murine melanoma cells. The CC50 dose of SG-GNPs against B16 cells were assessed with MTT assay and the anticancer activity was evaluated using Rhodamine 123, H2DCFDA and dual staining techniques. The induction of apoptosis by SG-GNPs against melanoma cells were confirmed with q-PCR analysis. The results of staining techniques prove that SG-GNPs increase the reactive oxygen species and decreased the mitochondrial membrane potential. It is further confirmed by the results of q-PCR analysis which shows increased apoptotic Bid, Bad, Casp3, Casp 9 genes and decreased antiapoptotic Bcl2 gene expression in SG-GNPs treated cells. Our results authentically prove the biosynthesized SG-GNPs induces apoptosis in melanoma cells and it possesses anticancer property.
    Keywords:  ; B16 cells; Nanoparticles; SG-GNPs; anticancer drug; melanoma
    DOI:  https://doi.org/10.1080/21691401.2019.1647224
  24. Am J Physiol Renal Physiol. 2019 Aug 07.
      OBJECTIVES: Diabetes could aggravate ischemia/reperfusion (I/R) injury, but the underlying mechanism is unclear. In this study, we aimed to investigate whether diabetes exacerbates renal I/R injury and its possible mechanism.MATERIALS AND METHODS: In vitro, HK-2 cells in the normal or high glucose (HG) condition were subjected to hypoxia 12h followed by reoxygenation 3h. The cell viability, intracellular adenosine triphosphate (ATP) content, mitochondrial membrane potential (MMP), reactive oxygen species (ROS) production and apoptosis were performed. In vivo, streptozotocin-induced diabetic and non-diabetic rats were subjected with I/R. Renal pathology, function, and apoptosis were evaluated by hematoxylin-eosin staining, transmission electron microscope (TEM) and western blot.
    RESULTS: Compared with normal glucose with hypoxia/reoxygenation (NH/R) group, mitochondrial function (ATP, MMP, ROS) and mitophagy were reduced in high glucose with H/R(HG+H/R), as well as expression of PINK1 and Parkin. Also, cells in HG+H/R group exhibited more apoptosis when compared with NH/R group through flow cytometry, TUNEL staining and Western blot. Compared with normal rats underwent I/R(NI/R), diabetic rats undergoing I/R(DI/R) exhibited more severe tubular damage and renal dysfunction, as well as expression of apoptotic protein Caspase-3. Meanwhile, diabetes alleviated mitophagy associated protein expression in rats subjected with I/R, including PINK1 and Parkin. TEM indicated that mitophagosome could be hardly observed and mitochondria morphology and structure were obviously damaged in DI/R group.
    CONCLUSION: Our results, for the first time, indicated that diabetes could aggravate I/R injury by repressing mitochondrial function and PINK1/Parkin-mediated mitophagy in vivo and in vitro.
    Keywords:  diabetes; mitochondrial function; mitophagy; renal ischemia reperfusion injury
    DOI:  https://doi.org/10.1152/ajprenal.00181.2019
  25. Cell Death Discov. 2019 ;5 125
      Breast cancer is the most common malignant tumor in women, and progress toward long-term survival has stagnated. Pristimerin, a natural quinonemethide triterpenoid, exhibits potential anti-tumor effects on various cancers. However, the underlying mechanism remains poorly understood. In this study, we found that pristimerin reduced the viability of breast cancer cells in vitro and the growth of xenografts in vivo, and these reductions were accompanied by thioredoxin-1 (Trx-1) inhibition and ASK1 and JNK activation. The results showed that pristimerin inhibited cell cycle progression and triggered cell apoptosis and autophagy. Furthermore, we found that the generation of reactive oxygen species (ROS) was a critical mediator in pristimerin-induced cell death. Enhanced ROS generation by pristimerin activated the ASK1/JNK signaling pathway. Inhibition of ROS with N-acetyl cysteine (NAC) significantly decreased pristimerin-induced cell death by inhibiting the phosphorylation of ASK1 and JNK. Taken together, these results suggest a critical role for the ROS/ASK1/JNK pathway in the anticancer activity of pristimerin.
    Keywords:  Breast cancer; Pharmacology
    DOI:  https://doi.org/10.1038/s41420-019-0208-0
  26. Mol Biol Rep. 2019 Aug 05.
      The purpose of this study was to investigate the effect of a superoxide-hydrogen peroxide (S-HP) imbalance of the superoxide dismutase manganese dependent (SOD2) gene, generated by paraquat and porphyrin exposure, on the keratinocytes cell line (HaCaT) oxidative metabolism. Paraquat acts increasing superoxide (O 2 ·- ) levels, while porphyrin increases hydrogen peroxide (H2O2) levels, acting as VV-SOD2-like and AA-SOD2-like molecules, respectively. First of all, HaCAT cells were treated with different concentrations of paraquat and porphyrin (1; 10; 30, and 70 μM) to determine the concentration of both that causes imbalance. After defining the concentration of paraquat and porphyrin (70 μM), a time curve was performed (1, 3, 6, and 24 h) to evaluate ROS production levels. Other oxidative parameters, such as nitric oxide (NO), lipoperoxidation (TBARS) and protein carbonyl, were evaluated after 24 h of incubation, as well as genotoxic analyses, apoptosis detection, and gene expression. Our findings revealed that paraquat exposure decreased cell viability, increasing lipoperoxidation, DNA damage, and apoptosis. On the other hand, porphyrin treatment increased cell viability and proliferation, ROS and NO production, triggering protein and DNA damage. In addition, porphyrin up-regulated Keap1 and Nrf2 gene expression, while paraquat decreased Nrf2 gene expression. In this sense, we suggested that the superoxide-hydrogen peroxide imbalance differentially modulates oxidative stress on keratinocytes cell line via Keap1-Nrf2 gene expression pathway.
    Keywords:  Keap1-Nrf2 pathway; Oxidative stress; Paraquat; Porphyrin; SOD2; Val16Ala-SOD2 SNP
    DOI:  https://doi.org/10.1007/s11033-019-05012-1
  27. Life Sci. 2019 Aug 06. pii: S0024-3205(19)30663-0. [Epub ahead of print] 116736
      AIMS: Diabetes mellitus (DM)-induced reproductive damage is an important cause of infertility for male DM patients, we herein evaluated the effects of catalpol on diabetic reproductive damage through the suppression of the AGEs/RAGE/Nox4 signaling pathway.METHODS: KK-Ay diabetic reproductive damage mice were administered with catalpol for 8 weeks, the testis/body weight ratio, testicular histopathology, the levels of endogenous hormone and the activity of testicular marker enzymes were determined. In vitro, the GC-2 cell injury model was induced by advanced glycation end-products (AGEs) and pretreated with catalpol. Cell viability, apoptosis, and oxidative stress markers were detected and the mechanism based on the AGEs/RAGE/Nox4 pathway was explored.
    KEY FINDINGS: Catalpol showed remarkable capacity on protecting diabetic reproductive damage by improving the histomorphology of the testes, increasing the testis/body weight ratio and activity of acid phosphatase (ACP), lactate dehydrogenase (LDH), gamma-glutamyl transferase (γ-GT). The reduced testosterone (T), luteinizing hormone (LH) and follicle-stimulating hormone (FSH) levels in DM mice were also reversed with catalpol intervention. Moreover, catalpol showed markedly effects of anti-oxidative in vivo and in vitro, which significantly down-regulated reactive oxygen species (ROS) levels and restored superoxide dismutase (SOD) activity, meanwhile decreased GC-2 cell apoptosis and Bax/Bcl-2 ratio. Moreover, the over-expression of receptors for AGEs (RAGE), NADPH oxidase type 4 (Nox4) and phosphorylation of nuclear transcription factor-κB p65 (NF-κB p65) were suppressed by catalpol.
    SIGNIFICANCE: Catalpol could alleviate DM-induced male reproductive damage by inhibiting oxidative stress-induced apoptosis and inflammation mediated by AGEs/RAGE/Nox4 signaling pathway.
    Keywords:  AGEs; Catalpol; Diabetes mellitus; Male reproductive damage; Nox4; RAGE
    DOI:  https://doi.org/10.1016/j.lfs.2019.116736
  28. Bioorg Chem. 2019 Jul 27. pii: S0045-2068(19)30496-1. [Epub ahead of print]91 103147
      Ugi reaction was a reliable procedure for the synthesis of new coumarin-quinoline frameworks. Excellent yields, mild reaction conditions and easily available and inexpensive starting materials are advantages of this protocol. Cytotoxic effects of fourteen products were investigated in A2780 human ovarian cancer cells. Two synthesized compounds (L11 and L12) exhibited more anti-cancer activity than other derivatives with IC50 values of 0.042 mmol/L and 0.102 mmol/L, respectively and were thus selected for further studies. Apoptosis was induced through the intrinsic pathway by activating caspase 9 and ended at the executioner pathway of caspase 3. Measurement of intracellular reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were also carried out for both of them. Further studies on a mechanism by Real Time-PCR and Western blot analysis were performed for anti-apoptotic proteins Bcl-2 and survivin both in mRNA and protein level relating to the untreated A2780 cells. The treatment of A2780 cells with compound L11 significantly (P-value ≤ 0.05) induced apoptosis by down-regulation of Bcl-2 and survivin both in mRNA and protein level via a single dose (0.042 mmol/L), as well as activation of caspase 9 and 3, loss of MMP, and high ROS. Accordingly, findings supported the first report under which the pro-apoptotic activity of compound L11 as an apoptosis-inducing agent was related to mitochondrial-mediated dysfunction signaling pathways. Molecular docking supports experimental outcomes. Evidently, coumarin-quinoline scaffolds are potentially favorable options for further assessment as influential chemotherapeutic agents for the future.
    Keywords:  Apoptosis; Bcl-2; Cancer; Coumarin-quinoline hybrids; Mitochondria-related pathways; Survivin
    DOI:  https://doi.org/10.1016/j.bioorg.2019.103147
  29. Mol Carcinog. 2019 Aug 08.
      A major concern in the clinical application of tumor necrosis factor related apoptosis-inducing ligand (TRAIL) in tumors is the development of resistance. Therefore, agents that can potentially restore TRAIL sensitivity are important therapeutic targets for cancer treatment. Herein, we evaluated lanatoside c and digoxin, both of which are widely used cardiac glycosides (CGs), for their ability to sensitize human hepatocellular carcinoma cells (Huh-7 and HepG2) through TRAIL-induced apoptosis. CGs functionalize TRAIL as shown by its effect on intracellular reactive oxygen species (ROS) generation, which damages mitochondrial integrity and thereby confers intrinsic apoptotic caspase cascade during combined treatment. Caspase activation is dependent on ROS as shown by the ability of CGs to generate ROS and the ROS-N-acetylcysteine (NAC) relationship, which inhibits apoptosis during cotreatment by preventing the formation of caspase-8 and -3. Furthermore, CGs triggered p38MAPK phosphorylation and NAC pre-exposure blocked p38MAPK phosphorylation, which demonstrated that p38MAPK was dependent upon ROS generation. Additionally, CGs were found to be potent inducers of AMPK-mediated protective autophagy as pharmacological and genetic autophagy inhibition reached the higher threshold of TRAIL-mediated apoptosis. Finally, CGs downregulated the expression of the antiapoptotic protein Bcl-2 and increased the translocation of proapoptotic protein cytochrome c, thereby inducing apoptosis. Collectively, these results indicate that CGs potentiate the enhanced cytotoxic capacity to TRAIL through ROS generation, p38MAPK phosphorylation, cell survival protein downregulation, and protective autophagy inhibition.
    Keywords:  ROS; TRAIL; apoptosis; autophagy; cardiac glycosides
    DOI:  https://doi.org/10.1002/mc.23096
  30. ACS Appl Mater Interfaces. 2019 Aug 06.
      Acute myocardial infarction (AMI), which can be extremely difficult to treat, is the worst deadly disease around the world. Reperfusion is expedient to reverse myocardial ischemia. However, during reperfusion, reactive oxygen species (ROS) produced by myocardial ischemia reperfusion injury (MIRI) and further cell apoptosis are the most serious challenge to cardiomyocytes. Therefore, searching for reagents that can simultaneously reduce oxidative damage and MIRI-induced apoptosis is the pivotal strategy to rescue injured cardiomyocytes. Nevertheless, current cardioprotective drugs have some shortcoming, such as cardiotoxicity, inadequate intravenous administration, or immature technology. Previous studies have shown that tetrahedral DNA nanostructures (TDNs) have biological safety with promising anti-inflammatory and antioxidative potential. However, the progress that TDNs have made in the biological behavior of cardiomyocytes has not been explored. In this experiment, a cellular model of MIRI was first established. Then, confirmed by a series of experiments, our study indicates that TDNs can significantly decrease oxidative damage and apoptosis by limiting the overexpression of ROS, along with effecting the expression of apoptosis-related proteins. In addition, western blot analysis demonstrated that TDNs could activate the Akt/Nrf2 signaling pathway to improve the myocardial injury induced by MIRI. Above all, the antioxidant and anti-apoptotic capacities of TDNs make them a potential therapeutic drug for MIRI. This study provides new ideas and directions for more homogeneous diseases induced by oxidative damage.
    DOI:  https://doi.org/10.1021/acsami.9b10645
  31. Poult Sci. 2019 Aug 03. pii: pez406. [Epub ahead of print]
      Mycoplasma gallisepticum (MG) infection produces a profound inflammatory response in the respiratory tract and evade birds' immune recognition to establish a chronic infection. Previous reports documented that the flavonoid baicalin possess potent anti-inflammatory, and antioxidant activities. However, whether baicalin prevent immune dysfunction is largely unknown. In the present study, the preventive effects of baicalin were determined on oxidative stress generation and apoptosis in the spleen of chickens infected with MG. Histopathological examination showed abnormal morphological changes including cell hyperplasia, lymphocytes depletion, and the red and white pulp of spleen were not clearly visible in the model group. Oxidative stress-related parameters were significantly (P < 0.05) increased in the model group. However, baicalin treatment significantly (P < 0.05) ameliorated oxidative stress and partially alleviated the abnormal morphological changes in the chicken spleen compared to model group. Terminal deoxynucleotidyl transferase-mediated dUTP nick endlabeling assay results, mRNA, and protein expression levels of mitochondrial apoptosis-related genes showed that baicalin significantly attenuated apoptosis. Moreover, baicalin restored the mRNA expression of mitochondrial dynamics-related genes and maintain the balance between mitochondrial inner and outer membranes. Intriguingly, the protective effects of baicalin were associated with the upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2)/Heme oxygenase-1 (HO-1) pathway and suppression of nuclear factor-kappa B (NF-κB) pathway in the spleen of chicken. In summary, these findings indicated that baicalin promoted mitochondrial dynamics imbalance and effectively prevents oxidative stress and apoptosis in the splenocytes of chickens infected with MG.
    Keywords:   Mycoplasma gallisepticum ; Nrf2/HO-1 pathway; apoptosis; baicalin; spleen
    DOI:  https://doi.org/10.3382/ps/pez406
  32. Biomed Pharmacother. 2019 Sep;pii: S0753-3322(19)31474-X. [Epub ahead of print]117 109156
      The present study was designed to study the protective effect of Ziziphora clinopodioides flavonoids (ZCF) against H2O2-induced oxidative stress in HUVEC cells. MTT assay was carried out to determine the cell viability of HUVEC cells following pretreatment with ZCF. Fluorescent microscopy measurements were performed to evaluate apoptosis of HUVEC cells. Furthermore, the effects of ZCF on the activities of antioxidants superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), malondialdehyde production (MDA) and lactic dehydrogenase (LDH) levels were analyzed. Apoptosis was observed by Hoechst33258 staining and AO staining. Real-time fluorescence quantitative polymerase chain reaction (Real-time PCR) was used to detect the expression of B-cell lymphoma/leukemia-2 (Bcl-2), Bcl-2-associated X protein (Bax) and aspartate proteolytic enzyme-3 (Caspase-3) mRNA. The expression of vascular endothelial growth factor receptor 2 (VEGFR2), protein kinase B (Akt), phosphorylated protein kinase B (p-Akt), Bax, Bcl-2 and Caspase-3 were detected by western blot. ZCF attenuated H2O2-induced cell death, as determined by the MTT assay. ZCF decreased malondialdehyde and lactic dehydrogenase levels, increased superoxide dismutase, glutathione peroxidase, catalase activities and inhibited apoptosis. Moreover, pretreatment with ZCF decreased the expression of Bax and Caspase-3 at mRNA level, increased the expression of Bcl-2 mRNA level, decreased the levels of VEGFR2, Bax and Caspase-3 protein, and increased the level of p-Akt / Akt and Bcl-2 protein in HUVEC cells. These results suggested that ZCF protected against H2O2-induced injury in HUVEC cells. The mechanism for this effect is related to the enhancement of antioxidant capacity, suppression of angiogenesis and apoptosis.
    Keywords:  Apoptosis; HUVEC cells; Hydrogen peroxide; Oxidative stress
    DOI:  https://doi.org/10.1016/j.biopha.2019.109156
  33. Hum Exp Toxicol. 2019 Aug 04. 960327119867758
      Carbon tetrachloride (CCL4) is often employed in the production of chlorofluorocarbons, petroleum refining, oil and rubber processing, and laboratory applications. Oral, subcutaneous, and inhalation exposure to CCL4 in animal studies have been shown to be capable of leading to various types of cancer (benign and malignant, liver, breast, and adrenal gland tumors). The present study also evaluated the protective role of infliximab (INF) against the deleterious effects of CCL4 on the intestinal system. Twenty-four male Sprague-Dawley rats were randomly assigned into three groups, control (n = 8), CCL4 (n = 8), and CCL4 + INF (n = 8). The control group received 1 mL isotonic saline solution only via intraperitoneal (i.p.) injection. The CCL4 group received a single i.p. dose of 2 mL/kg CCL4. The CCL4 + INF group received a single i.p. dose of 7 mg/kg INF followed 24 h later by a single dose of 2 mL/kg CCL4. All rats were euthanized 2 days following drug administration. CCL4 group samples also exhibited diffuse loss of enterocytes, vascular congestion, neutrophil infiltration, an extension of the subepithelial space and significant epithelial lifting along the length of the villi with a few denuded villous tips. In addition, CCL4 treatment increased intestinal malondialdehyde (MDA) level and caspase-3 positivity. On the other hand, INF decreased MDA levels, caspase-3 positivity, and loss of villous. Our findings suggest that CCL4 appears to exert a highly deleterious effect on the intestinal mucosa. On the other hand, INF is effective in preventing this CCL4-induced intestinal injury by reducing oxidative stress and apoptosis.
    Keywords:  Apoptosis; carbon tetrachloride; infliximab; lipid peroxidation; rat; small intestine
    DOI:  https://doi.org/10.1177/0960327119867758
  34. Cell Microbiol. 2019 Aug 06. e13094
      Mycobacterium avium, a slow-growing non-tuberculosis mycobacterium, causes fever, diarrhoea, loss of appetite, and weight loss in immunocompromised people. We have proposed that endoplasmic reticulum (ER) stress-mediated apoptosis plays a critical role in removing intracellular mycobacteria. In the present study, we investigated the role of the regulated IRE1-dependent decay (RIDD) pathway in macrophages during M. avium infection based on its role in the regulation of gene expression. The inositol-requiring enzyme 1 (IRE1)/apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) signalling pathway was activated in macrophages after infection with M. avium. The expression of RIDD-associated genes, such as Bloc1s1 and St3gal5, was decreased in M. avium-infected macrophages. Interestingly, M. avium-induced apoptosis was significantly suppressed by pre-treatment with irestatin (inhibitor of IRE1α) and 4μ8c (RIDD blocker). Macrophages pre-treated with N-acetyl cysteine (NAC) showed decreased levels of reactive oxygen species (ROS), IRE1α, and apoptosis after M. avium infection. The expression of Bloc1s1 and St3gal5 was increased in NAC-pre-treated macrophages following infection with M. avium. Growth of M. avium was significantly increased in irestatin-, 4μ8c-, and NAC-treated macrophages compared to the control. The data indicate that the ROS-mediated ER stress response induces apoptosis of M. avium-infected macrophages by activating IRE1α-RIDD. Thus, activation of IRE1α suppresses the intracellular survival of M. avium in macrophages.
    Keywords:  Apoptosis; ER stress; IRE1α; Mycobacterium avium; RIDD
    DOI:  https://doi.org/10.1111/cmi.13094
  35. Biomed Pharmacother. 2019 Aug 01. pii: S0753-3322(18)38322-7. [Epub ahead of print]118 108946
      Prostate cancer is one of the most frequently diagnosed neoplasms among men in the world. However, molecular mechanisms underlying the progression of prostate cancer are still unclear. In the study, we investigated the effects of ubiquitin specific protease 17 (USP17) on prostate cancer growth. The results indicated that USP17 expression was markedly increased in prostate cancer tissues and cell lines. Repressing USP17 expression significantly reduced the proliferation, migration and invasion of prostate cancer cells using cell counting kit-8 (CCK-8), colony formation and transwell assays. In addition, apoptosis was significantly induced by USP17 knockdown via increasing the expression of cleaved Caspase-9/-3 and poly (ADP)-ribose polymerase (PARP), as well as Cyto-c. Further, USP17 silence evidently promoted reactive oxygen species (ROS) production in prostate cancer cells. Nuclear nuclear factor-κB (NF-κB)/p65 expression and total NF-κB/p65 phosphorylation were markedly down-regulated by USP17 repression. Intriguingly, blocking ROS generation using its scavenger of N-acetyl-l-cysteine (NAC) significantly abrogated USP17 knockdown-induced apoptosis and -inhibited NF-κB/p65 signaling in vitro. Our data also showed that USP17 silence impaired tumor growth in the subcutaneous mouse model in vivo. Taken together, our results suggested that USP17 decrease might exert anti-tumor activities against prostate cancer growth by inducing apoptosis and suppressing NF-κB/p65 signaling via the promotion of ROS. Thus, USP17 could be served as a promising candidate to develop effective therapeutic strategy against prostate cancer progression.
    Keywords:  Apoptosis; NF-κB/p65; Prostate cancer; ROS; USP17
    DOI:  https://doi.org/10.1016/j.biopha.2019.108946
  36. J Biomed Nanotechnol. 2019 Sep 01. 15(9): 1867-1880
      The present study aims to evaluate the effect of the ethyl acetate extract of Cichorium (EAEC) as a novel photosensitizer in photodynamic therapy (PDT) of colorectal carcinoma (CRC) HCT116 and SW620 cells. The absorption and fluorescence spectra of EAEC were measured using a UV-vis spectrophotometer and fluorescence spectrophotometer, respectively. EAEC-induced reactive oxygen species (ROS) production in HCT116 and SW620 cells was detected using 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) and glutathione/glutathione disulfide (GSH/GSSG). The photo- and dark toxicities of EAEC were estimated using the Cell Counting Kit-8 (CCK-8) assay. Cellular uptake and localization of EAEC were detected by confocal laser fluorescence microscopy. Annexin V-FITC/PI staining, Western blotting and immunofluorescence staining were used to assess apoptosis and autophagy. The antitumor activity of EAEC was confirmed in a xenograft model. Finally, effects on the PERK pathway were verified using qRT-PCR and Western blotting. EAEC displayed absorption and fluorescence emission peaks at 660 nm and 678 nm, respectively. EAEC induced ROS production in CRC cells. Assessment of dark toxicity showed that treatment with EAEC alone induced little cytotoxicity in CRC or normal cells but that EAEC-PDT induced significant photocytotoxicity in CRC cells in a time- and dose-dependent manner. After cellular uptake, EAEC was located in the mitochondria. Treatment with EAEC-PDT reduced xenograft tumor size. Further evaluation suggested that activation of the PERK pathway mediates these effects, as the apoptotic rate and autophagy flux increased markedly after EAEC-PDT. EAEC, a natural photosensitizer extracted from Cichorium, displays potential utility in PDT of CRC by targeting the PERK pathway.
    DOI:  https://doi.org/10.1166/jbn.2019.2825
  37. Mol Biol Rep. 2019 Aug 07.
      Neuroprotection using compounds with dual functions of anti-apoptotic and antioxidant effects fight against neurodegeneration. Vitamin K2 acts as a cofactor in many biochemical pathways, including sphingolipid synthesis in the nervous system, which is involved in many cellular events, including proliferation, differentiation, cellular communication, and alteration. This study aimed to investigate the protective effects of vitamin K2 in PC12 cells as an in vitro model of Parkinson's disease. The protective effects of vitamin K2 against 6-OHDA-induced apoptosis in PC12 cells were assessed using resazurin for viability, DCF-DA for ROS level, DTNB for glutathione level, flow cytometry for sub G1, and western blot analysis for detecting bax and pro-caspase-3 expression level. The results showed that 6-OHDA significantly decreased cell viability, glutathione and pro-caspase-3 levels, and increased ROS, the amount of bax in PC12 cells, while the pretreatment with 5 μM vitamin K2 significantly decreased the cell death induced by 6-OHDA. Generally, the results may present a new insight about the potential protective action of vitamin K2 against the progression of Parkinson's disease. Further studies may warrant the use of vitamin K2 as an antioxidant and anti-apoptotic agent in slowing nerve injury in neurodegenerative disease, particularly in Parkinson's disease.
    Keywords:  6-OHDA; Apoptosis; PC12; Parkinson’s disease; Vitamin K2
    DOI:  https://doi.org/10.1007/s11033-019-05011-2
  38. Biomed Pharmacother. 2019 Aug 03. pii: S0753-3322(19)32344-3. [Epub ahead of print]118 109246
      Icariin (ICA) and phosphorylated icariin (pICA) have excellent antiviral and antioxidant effects. However, whether ICA and pICA cause anti-LPS-induced intestinal damage remains unclear. In this study, we used Caco-2 cells as a model to investigate the protective effects of ICA and pICA on human colonic epithelial cells and explore their potential mechanisms. Our results indicated that ICA and pICA increased cell viability and decreased lactate dehydrogenase activity in Caco-2 cells. ICA and pICA also attenuated LPS-induced changes in intestinal epithelial cell permeability and reduced the levels of oxidative stress indicators, such as reactive oxygen species, malondialdehyde, and hydrogen peroxide, in Caco-2 cells. Antioxidant indicators, such as superoxide dismutase, glutathione peroxidase, catalase and total antioxidant capacity, were increased, while the levels of IL-1β, IL-6, IL-8 and TNF-α were reduced in the ICA and pICA groups. Furthermore, ICA and pICA decreased the gene abundance and enzyme activities of caspase-3, -8, -9 and -10 in Caco-2 cells. Our data suggest that ICA and pICA effectively attenuated LPS-induced changes in the oxidative stress, inflammation, apoptosis and intestinal permeability of intestinal epithelial cells. These findings provide new insight for treating LPS-induced intestinal injury.
    Keywords:  Caco-2 cells; Icariin; Intestinal injury; LPS; Phosphorylated icariin
    DOI:  https://doi.org/10.1016/j.biopha.2019.109246
  39. J Cell Physiol. 2019 Aug 07.
      Hypoxia is a common pathological process caused by insufficient oxygen. Long noncoding RNAs (lncRNAs) have been proven to participate in this pathology. Hypoxia is reported to significantly reduce the secretion of tissue inhibitor of metalloproteinase 2 (TIMP2) and TIMP2 induces pheochromocytoma-12 (PC12) cell cycle arrest. Thus, our study aimed to explore the mechanism by which lncRNA maternally expressed gene 3 (MEG3) was implicated in hypoxia-induced PC12 cell injury through TIMP2 promoter methylation. To elucidate the potential biological significance of MEG3 and the regulatory mechanism between MEG3 and TIMP2, a hypoxia-induced PC12 cell injury model was generated. The hypoxia-exposed cells were subjected to a series of overexpression plasmids and short hairpin RNAs, followed by the measurement of levels of MEG3, TIMP2, lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD), reactive oxygen species (ROS), Bcl-2-associated X protein, B-cell lymphoma-2, and caspase-3, as well as the changes in MMP, cell proliferation, apoptosis, and cell cycle progression. On the basis of the findings, MEG3 was upregulated in hypoxia-injured PC12 cells. MEG3 recruited methylation proteins DNMT3a, DNMT3b, and MBD1 and accelerated TIMP2 promoter methylation, which in turn inhibited its expression. Moreover, PC12 cells following MEG3 silencing and TIMP2 overexpression exhibited significantly decreased levels of LDH, MDA, and ROS along with cell apoptosis, yet increased SOD and MMP levels, as well as cell cycle entry to the S phase and cell proliferation. In conclusion, MEG3 silencing suppresses hypoxia-induced PC12 cell injury by inhibiting TIMP2 promoter methylation. This study may provide novel therapeutic targets for hypoxia-induced injury.
    Keywords:  MEG3; TIMP2; hypoxic injury; long noncoding RNA; pheochromocytoma-12 cell; promoter methylation
    DOI:  https://doi.org/10.1002/jcp.29085
  40. PLoS One. 2019 ;14(8): e0220581
      Sirt1, a member of the sirtuin gene family, encodes the most conserved mammalian NAD+-dependent deacetylase enzyme responsible for removing acetyl groups from many proteins. The Sirt1 gene is known as a longevity gene whose knockout in mice leads to decreased lifespan relative to the wild type. This study aimed to explore phenotypic changes in zebrafish Sirt1-knockouts and to investigate the function of the Sirt1 gene. Targeted knockout of Sirt1 in zebrafish (Danio rerio) was achieved using the CRISPR-Cas9 genome editing system. We created a 4-bp insertion-homozygote Sirt1-knockout zebrafish. Although there was no evident difference in appearance in the early stages of development, a significant increase in reactive oxygen species and in the extent of apoptosis in Sirt1-knockout zebrafish was observed. Sirt1 knockout caused inflammatory genes, including IL-1b, IL-6 and TNF-α to be highly expressed. Additionally, the lack of Sirt1 caused chronic inflammation and intestinal atrophy, thereby increasing pro-apoptotic events, which ultimately reduced the lifespan of transgenic zebrafish. Overall, our data demonstrate that lack of Sirt1 caused a significantly increased generation of reactive oxygen species that resulted in chronic inflammation and regeneration. Continuous repetition of these events played an important role in accelerating aging, thereby decreasing lifespan. Our findings using the knockout zebrafish model confirmed the association of the Sirt1 gene to aging processes and lifespan. Furthermore, the Sirt1-knockout mutant zebrafish developed in our study will surely be a valuable model to explore the mechanism of chronic inflammation.
    DOI:  https://doi.org/10.1371/journal.pone.0220581
  41. Free Radic Biol Med. 2019 Aug 06. pii: S0891-5849(19)30778-6. [Epub ahead of print]
      Postovulatory aging is known to compromise the oocyte quality as well as subsequent embryo development in many different animal models, and becomes one of the most intractable issues that limit the outcome of human assisted reproductive technology (ART). However, the strategies to prevent the deterioration of aged oocytes and relevant mechanisms are still underexplored. Here, we find that supplementation of CoQ10, a natural antioxidant present in human follicular fluids, is able to restore the postovulatory aging-induced fragmentation of oocytes and decline of fertilization. Importantly, we show that CoQ10 supplementation recovers postovulatory aging-caused meiotic defects such as disruption of spindle assembly, misalignment of chromosome, disappearance of actin cap, and abnormal distribution patterns of mitochondria and cortical granules. In addition, CoQ10 protects aged oocytes from premature exocytosis of ovastacin, cleavage of sperm binding site ZP2, and loss of localization of Juno, to maintain the fertilization potential. Notably, CoQ10 suppresses the aging-induced oxidative stress by reducing the levels of superoxide and DNA damage, ultimately inhibiting the apoptosis. Taken together, our findings demonstrate that CoQ10 supplementation is a feasible and effective way to prevent postovulatory aging and preserve the oocyte quality, potentially contributing to improve the successful rate of IVF (in vitro fertilization) and ICSI (intracytoplasmic sperm injection) during human ART.
    Keywords:  Apoptosis; CoQ10; DNA damage; Oocyte quality; Oxidative stress; Postovulatory aging
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2019.08.002
  42. Biosci Rep. 2019 Aug 05. pii: BSR20190109. [Epub ahead of print]
      Diabetes mellitus is a potential etiology of disc degeneration. Glucagon-like peptide-1 (GLP-1) is currently regarded as a powerful treatment option for type 2 diabetes. Apart from the beneficial effects on glycemic control, GLP-1 has been reported to exert functions in a variety of tissues on modulation of cell proliferation, differentiation, and apoptosis. However, little is known regarding the effects of GLP-1 on nucleus pulposus cells (NPCs). In the present study, we investigated the effects of liraglutide (LIR), a long-lasting GLP-1 analogue, on apoptosis of human NPCs and the underlying mechanisms involved. We confirmed the presence of GLP-1 receptor (GLP-1R) in NPCs. Our data demonstrated that liraglutide inhibited the apoptosis of NPCs induced by high glucose, as detected by Annexin V/PI and ELISA assays. Moreover, liraglutide downregulated caspase-3 activity at intermediate concentration (100 nM) for maximum effect. Further analysis suggested that liraglutide suppressed reactive oxygen species (ROS) generation and stimulated the phosphorylation of Akt under high glucose condition. Pretreatment of cells with the Phosphoinositide 3-kinase (PI3K) inhibitor LY294002 and siRNAs GLP-1R abrogated the liraglutide-induced activation of Akt and the protective effects on NPCs apoptosis. In conclusion, liraglutide could directly protect NPCs against high glucose-induced apoptosis by inhibiting oxidative stress and activating the PI3K/Akt/caspase-3 signaling pathway via GLP-1R.
    Keywords:  apoptosis; liraglutide; nucleus pulposus cells; oxidative stress; signaling pathway
    DOI:  https://doi.org/10.1042/BSR20190109
  43. Eur Rev Med Pharmacol Sci. 2019 Aug;pii: 18647. [Epub ahead of print]23(3 Suppl): 192-200
      OBJECTIVE: To explore the influences of high glucose on the proliferation and apoptosis of prostate cancer cells and analyze its possible mechanism of action.MATERIALS AND METHODS: Human prostate cancer cell line LNCaP was divided into control group, mannitol group, and high glucose group. Then, the proliferation in each group was detected via methyl-thiazolyl-tetrazolium (MTT) assay. Hoechst staining assay was performed to determine the apoptosis level in each group. Western blotting was employed to measure the expression levels of apoptosis-related proteins and nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and γ-glutamylcysteine synthetase (γ-GCS) proteins. The cellular reactive oxygen species (ROS) level was measured through 2,7-dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay. Enzyme-linked immunosorbent assay (ELISA) was carried out to detect the content of lactate dehydrogenase (LDH) and inflammatory factors.
    RESULTS: High glucose significantly promoted the proliferation of prostate cancer cells LNCaP (p<0.01) and increased the apoptosis level of cells (p<0.01). In high glucose group, the expression level of Caspase-3 protein was overtly increased (p<0.01), while that of B-cell lymphoma-2 (Bcl-2)/Bcl-2 associated X protein (Bax) was significantly decreased (p<0.01). High glucose group had clearly increased the content of ROS (p<0.01), LDH (p<0.01), and interleukin-6 (IL-6) (p<0.01), but decreased the content of IL-10 (p<0.01). High glucose notably lowered the protein expression levels of Nrf2, HO-1, and γ-GCS in the cells (p<0.01).
    CONCLUSIONS: High glucose represses the activation of the Nrf2/anti-oxidation response element (ARE) signaling pathway in prostate cancer cells and increases the content of ROS, IL-6, and the expression of apoptotic proteins in the cells, thus promoting the apoptosis of prostate cancer cells.
    DOI:  https://doi.org/10.26355/eurrev_201908_18647
  44. Chemosphere. 2019 Jul 31. pii: S0045-6535(19)31725-4. [Epub ahead of print]237 124501
      Several evidences from the literature showed that the coexistence of nickel and zinc in polluted waters is related to the similarity in their geogenic and anthropogenic factors. Although most environmental exposures to metals do not occur singly, there is a paucity of scientific knowledge on the effects of zinc and nickel co-exposure on mammalian reproductive health. The present study investigated the influence of co-exposure to nickel and zinc on male reproductive function in rats. Experimental rats were co-exposed to environmentally relevant concentrations of waterborne nickel (75 and 150 μg NiCl2 L-1) and zinc (100 and 200 μg ZnCl2 L-1) for 45 successive days. Subsequently, reproductive hormones were assayed whereas the hypothalamus, epididymis and testes of the rats were processed for the assessment of oxidative stress and inflammation indices, caspase-3 activity and histology. Results indicated that co-exposure to nickel and zinc significantly (p < 0.05) abolished nickel-mediated diminution of antioxidant defense mechanisms while diminishing levels of reactive oxygen and nitrogen species and lipid peroxidation in the hypothalamus, epididymis and testes of the exposed rats. Additionally, co-exposure to zinc abated nickel-mediated diminutions in luteinizing hormone, follicle-stimulating hormone, serum and intra-testicular testosterone with concomitant enhancement of sperm production and quality. Further, zinc abrogated nickel-mediated elevation in inflammatory biomarkers including nitric oxide, tumor necrosis factor alpha, interleukin-1 beta as well as caspase-3 activity. The protective influence of zinc on nicked-induced reproductive toxicity was well supported by histological data. Overall, zinc ameliorated nickel-induced reproductive dysfunction via its anti-oxidant, anti-inflammatory, anti-apoptotic and spermato-protective activities in rats.
    Keywords:  Caspase-3; Male reproduction; Nickel; Oxido-inflammation; Zinc
    DOI:  https://doi.org/10.1016/j.chemosphere.2019.124501
  45. Med Sci Monit. 2019 Aug 05. 25 5833-5840
      BACKGROUND Salidroside, a natural dietary isothiocyanate, has been widely studied for its multiple effects, including promoting proliferation, anti-inflammation, and anti-apoptosis. In the present study, these effects of Salidroside were explored to assess whether it could prevent osteoarthritis (OA) in vitro. MATERIAL AND METHODS The cytotoxic and proliferating effects of Salidroside on chondrocytes were detected by use of the Cell Counting Kit 8 assay. The expression levels of proteins were detected by Western blot. The cell apoptosis level was assessed by flow cytometry, and the levels of ROS, NO, caspase 3, and caspase 9 were assessed to evaluate the level of apoptosis. The expression level of pro-inflammatory factors was detected by ELISA. RESULTS Our results demonstrated that Salidroside promotes chondrocytes proliferation, inhibits IL-1ß-induced apoptosis and inflammation, and scavenges reactive oxygen species (ROS) and NO of chondrocytes. Salidroside upregulates the level of Bcl-2 and downregulates the level of Bax. Salidroside also inhibits the production of caspase 3/9 and suppresses the phosphorylation of PI3K and AKT. CONCLUSIONS Our results suggest that Salidroside prevents OA by its powerful pro-proliferating, anti-phlogistic, and anti-apoptotic effects by inhibiting PI3K/AKT.
    DOI:  https://doi.org/10.12659/MSM.917851
  46. J Cell Mol Med. 2019 Aug 05.
      Myocardial ischaemia/reperfusion (I/R) injury attenuates the beneficial effects of reperfusion therapy. Poly(ADP-ribose) polymerase (PARP) is overactivated during myocardial I/R injury. Mitophagy plays a critical role in the development of myocardial I/R injury. However, the effect of PARP activation on mitophagy in cardiomyocytes is unknown. In this study, we found that I/R induced PARP activation and mitophagy in mouse hearts. Poly(ADP-ribose) polymerase inhibition reduced the infarct size and suppressed mitophagy after myocardial I/R injury. In vitro, hypoxia/reoxygenation (H/R) activated PARP, promoted mitophagy and induced cell apoptosis in cardiomyocytes. Poly(ADP-ribose) polymerase inhibition suppressed H/R-induced mitophagy and cell apoptosis. Parkin knockdown with lentivirus vectors inhibited mitophagy and prevented cell apoptosis in H/R-treated cells. Poly(ADP-ribose) polymerase inhibition prevented the loss of the mitochondrial membrane potential (ΔΨm). Cyclosporin A maintained ΔΨm and suppressed mitophagy but FCCP reduced the effect of PARP inhibition on ΔΨm and promoted mitophagy, indicating the critical role of ΔΨm in H/R-induced mitophagy. Furthermore, reactive oxygen species (ROS) and poly(ADP-ribosylation) of CypD and TSPO might contribute to the regulation of ΔΨm by PARP. Our findings thus suggest that PARP inhibition protects against I/R-induced cell apoptosis by suppressing excessive mitophagy via the ΔΨm/Parkin pathway.
    Keywords:  cell apoptosis; mitochondrial membrane potential; mitophagy; poly(ADP-ribose) polymerase; poly-ADP-ribosylation
    DOI:  https://doi.org/10.1111/jcmm.14573
  47. Int J Mol Sci. 2019 Aug 05. pii: E3820. [Epub ahead of print]20(15):
      Licochalcone A (LCA) is a chalcone that is predominantly found in the root of Glycyrrhiza species, which is widely used as an herbal medicine. Although previous studies have reported that LCA has a wide range of pharmacological effects, evidence for the underlying molecular mechanism of its anti-cancer efficacy is still lacking. In this study, we investigated the anti-proliferative effect of LCA on human bladder cancer cells, and found that LCA induced cell cycle arrest at G2/M phase and apoptotic cell death. Our data showed that LCA inhibited the expression of cyclin A, cyclin B1, and Wee1, but increased the expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1, and increased p21 was bound to Cdc2 and Cdk2. LCA activated caspase-8 and -9, which are involved in the initiation of extrinsic and intrinsic apoptosis pathways, respectively, and also increased caspase-3 activity, a typical effect caspase, subsequently leading to poly (ADP-ribose) polymerase cleavage. Additionally, LCA increased the Bax/Bcl-2 ratio, and reduced the integrity of mitochondria, which contributed to the discharge of cytochrome c from the mitochondria to the cytoplasm. Moreover, LCA enhanced the intracellular levels of reactive oxygen species (ROS); however, the interruption of ROS generation using ROS scavenger led to escape from LCA-mediated G2/M arrest and apoptosis. Collectively, the present data indicate that LCA can inhibit the proliferation of human bladder cancer cells by inducing ROS-dependent G2/M phase arrest and apoptosis.
    Keywords:  G2/M arrest; Licochalcone A; ROS; apoptosis; bladder cancer
    DOI:  https://doi.org/10.3390/ijms20153820
  48. J Exp Clin Cancer Res. 2019 Aug 05. 38(1): 338
      BACKGROUND: The GKN2 is a secretory protein, whose levels decrease in gastric cancer. The present study aimed to investigate the expression, function and mechanism of action of GKN2 in gastric cancer.METHODS: Molecular biology assays were performed to elucidate the function and underlying mechanisms of GKN2 in gastric cancer under stress-induced condition in vivo and in vitro. Clinical specimens were used to assess the correlation of GKN2 and prognosis.
    RESULTS: We found that overexpression of GKN2 significantly enhanced apoptosis and growth arrest in vitro. GKN2 expression increased in gastric cancer cells exposed to hydrogen peroxide and promoted reactive oxygen species-induced mitochondrial dysfunction and resulted in increased cell apoptosis via inhibition of NF-κB signaling pathway and activation of JNK signaling pathway through the direct interaction of GKN2 with Hsc70. Trefoil factor 1 might contribute to the tumor suppressing effects of GKN2. MiR-216a downregulated GKN2 expression. GKN2 also inhibited xenograft tumor growth and was an independent and significant prognostic factor for patients with gastric cancer treated with oxaliplatin.
    CONCLUSIONS: Taken together, our data indicate that GKN2 may increase sensitivity of GC cells to the drugs which increase ROS levels in tumors. Inhibition of the interaction between GKN2 and Hsc70 could attenuate the effects induced by GKN2. GKN2 overexpression could be used to determine the subgroup of patients to obtain the more favorable outcome of oxaliplatin treatment and may be used as biomarker of the prognosis of this cancer.
    Keywords:  Apoptosis; GKN2; Gastric cancer; Oxidative stress
    DOI:  https://doi.org/10.1186/s13046-019-1336-3
  49. J Bioenerg Biomembr. 2019 Aug 06.
      HESA-A is an herbal-marine compound which improves the quality of life of end-stage cancer patients. The aim of the present study was to evaluate the possible protective effect of HESA-A against IR-induced genotoxicity and apoptosis in rat bone marrow. Rats were given HESA-A orally at doses of 150 and 300 mg/kg body weight for seven consecutive days. On the seventh day, the rats were irradiated with 4 Gy X-rays at 1 h after the last oral administration. The micronucleus assay, reactive oxygen species (ROS) level analysis, hematological analysis and flow cytometry were used to assess radiation antagonistic potential of HESA-A. Administration of 150 and 300 mg/kg of HESA-A to irradiated rats significantly reduced the frequencies of micronucleated polychromatic erythrocytes (MnPCEs) and micronucleated normochromatic erythrocytes (MnNCEs), and also increased PCE/(PCE + NCE) ratio in bone marrow cells. Moreover, pretreatment of irradiated rats with HESA-A (150 and 300 mg/kg) significantly decreased ROS level and apoptosis in bone marrow cells, and also increased white blood cells count in peripheral blood. For the first time in this study, it was observed that HESA-A can have protective effects against radiation-induced genotoxicity and apoptosis in bone marrow cells. Therefore, HESA-A can be considered as a candidate for future studies to reduce the side effects induced by radiotherapy in cancer patients.
    Keywords:  Bone marrow cells; Genotoxicity; HESA-A; Ionizing radiation; Radioprotector
    DOI:  https://doi.org/10.1007/s10863-019-09808-5
  50. Comp Biochem Physiol C Toxicol Pharmacol. 2019 Aug 06. pii: S1532-0456(19)30301-1. [Epub ahead of print] 108585
      Compound ammonium glycyrrhizin (CAG) protects hepatocytes from injury induced by lipopolysaccharide (LPS)/florfenicol (FFC) through a mitochondrial pathway. On this basis, the research was aimed to investigate whether CAG protects hepatocytes from injury induced by LPS/FFC through oxidative stress and the MAPK pathway. For liver injury induced by LPS/FFC, not only CAG can protect hepatocytes and prevent membrane permeability from being increased, but also the activities of ALT and AST were decreased significantly by CAG. Flow cytometry analysis indicated that the apoptosis rate (35.65 ± 2.48%) of LPS/FFC group was significantly higher than that of the control group (8.60 ± 0.32%). CAG (concentration of 0.01 μg/mL, 0.1 μg/mL, 1 μg/mL) significantly decreased the apoptosis rate (23.69 ± 0.54%, 14.92 ± 2.45% and 9.47 ± 1.28%) for the liver injury induced by LPS/FFC. The activities of SOD and GSH were increased with the increased concentration of CAG, and the activity of MDA was decreased with the increased concentration of CAG. All the mRNA and proteins expression levels were increased by LPS/FFC-induced liver injury which associated with the MAPK pathway, and those of the CAG group were decreased with the increased concentration of CAG. And the change of caspase-3 activity was consistent with that of proteins and mRNA. It is suggested that LPS/FFC can induce liver injury through apoptosis and the CAG can protect hepatocytes from injury through the MAPK pathway and oxidative stress.
    Keywords:  Apoptosis; Compound ammonium glycyrrhizin; Liver injury; Oxidative stress; The MAPK pathway
    DOI:  https://doi.org/10.1016/j.cbpc.2019.108585
  51. J Physiol Biochem. 2019 Aug 10.
      Preeclampsia (PE) is a hypertensive disease associated with increased endothelial cell dysfunction caused by systemic oxidative stress. Alpha-actinin-4 (ACTN4) is a member of the α-actinin family of actin crosslinking proteins that are upregulated in several types of cancer. However, its role in PE remains unclear. In this study, we found that ACTN4 was localized in placenta vascular endothelial cells (ECs), and its expression was downregulated in primary human umbilical vein endothelial cells (HUVECs) from severe preeclamptic patients compared to that in HUVECs from normotensive pregnant women. ACTN4 expression was also decreased in normotensive HUVECs treated with H2O2. Downregulation of ACTN4 by siRNA or H2O2 treatment promoted normotensive HUVEC apoptosis and increased p38-MAPK phosphorylation along with elevated levels of p53 phosphorylation, caspase cascade proteins, and bax and repressed expression of bcl-2. Conversely, upregulation of ACTN4 in PE HUVECs significantly inhibited apoptosis and decreased p38-MAPK phosphorylation compared to that of the PE HUVEC controls. In addition, overexpression of ACTN4 in normotensive HUVECs attenuated H2O2 treatment-induced apoptosis with decreased p53 phosphorylation, caspase cascade, and bax expression levels and increased expression of bcl-2 compared to that of only H2O2 treatment. Moreover, the suppression of ACTN4 induced apoptosis, which could be blocked by the p38-MAPK inhibitor SB202190. Collectively, these results demonstrate that dysregulated ACTN4 expression may be associated with PE due to its effects on endothelial cell apoptosis via the p38-MAPK/p53 apoptosis pathway.
    Keywords:  Apoptosis; Endothelial cell dysfunction; Oxidative stress; P38-MAPK/p53 pathway; Preeclampsia
    DOI:  https://doi.org/10.1007/s13105-019-00700-9
  52. Cell Death Dis. 2019 Aug 07. 10(8): 593
      Colorectal cancer (CRC) is a common malignant gastrointestinal tumor with high mortality worldwide. Drug resistance and cytotoxicity to normal cells are the main causes of chemotherapeutic treatment failure in CRC. Therefore, extracting the bioactive compounds from natural products with anti-carcinogenic activity and minimal side-effects is a promising strategy against CRC. The present study aims to evaluate the anti-carcinogenic properties of avenanthramides (AVNs) extracted from oats bran and clarify the underlying molecular mechanisms. We demonstrated that AVNs treatment suppressed mitochondrial bioenergetic generation, resulting in mitochondrial swelling and increased reactive oxygen species (ROS) production. Further study indicated that AVNs treatment significantly reduced DDX3 expression, an oncogenic RNA helicase highly expressed in human CRC tissues. DDX3 overexpression reversed the ROS-mediated CRC apoptosis induced by AVNs. Of note, we identified Avenanthramide A (AVN A) as the effective ingredient in AVNs extracts. AVN A blocked the ATPase activity of DDX3 and induced its degradation by directly binding to the Arg287 and Arg294 residues in DDX3. In conclusion, these innovative findings highlight that AVNs extracts, in particular its bioactive compound AVN A may crack the current hurdles in the way of CRC treatment.
    DOI:  https://doi.org/10.1038/s41419-019-1825-5
  53. Biomed Pharmacother. 2019 Sep;pii: S0753-3322(19)31589-6. [Epub ahead of print]117 109177
      Exposure to ambient fine particulate matter (fine PM) pollution has been previously associated with ocular surface diseases. But, to the best of our knowledge, the in vivo long-term effects of fine PM on the ocular surface have not been investigated. We aimed to evaluate the effects of fine PM on cultured human corneal epithelial (HCE) cells and on the ocular surfaces of mice, with standard reference material of fine PM(SRM 2786). We applied fine PM suspension to the eyes of C57BL/6 mice for up to 6 months. In vivo examinations, including tear secretion, tear film break-up time (TBUT) and corneal fluorescein staining, were performed in the 3rd and 6th month. At the end of the in vivo study, the corneal histological changes and conjunctival goblet cells were examined by staining, and cytokines in tissue were also detected. In addition, HCE cells were treated with fine PM for 12 h and 24 h. Then, cell apoptosis and reactive oxygen species (ROS) formation was detected. We found that fine PM damages the mouse eye in a dose- and time-dependent manner. In mice, the tear secretion and tear film break-up time were significantly reduced, along with the development of corneal epithelial damage, apoptosis of conjunctival epithelial cells and hypoplasia of conjunctival goblet cells. In addition, IL-18, IL-22, IL-23 and MCP-1 were increased in both conjunctiva and cornea of the fine PM-treated animals. Furthermore, increased apoptosis and ROS production were observed in time- and dose-dependent manner in HCE cells after fine PM exposure for 12 h and 24 h. Our results indicate that fine PM is cytotoxic to both HCE cells and the ocular surface. Long-term topical application of fine PM suspension in mice results in ocular surface changes that are similar to those observed with dry eye.
    Keywords:  Conjunctiva; Corneal epithelial cell; Fine particulate matter; Ocular surface; Tear film break-up time
    DOI:  https://doi.org/10.1016/j.biopha.2019.109177
  54. Molecules. 2019 Aug 05. pii: E2839. [Epub ahead of print]24(15):
      Ozone is the most oxidant tropospheric pollutant gas, causing damage through the formation of reactive oxygen and nitrogen species. Reactive species induce the nuclear factor-kappa B (NF-κB) activation leading to neuroinflammation characterized by astrocytosis, microgliosis, and apoptotic cell death. There is interest in evaluating the pharmacological activity of natural antioxidants to confer neuroprotection against the damage caused by ozone in highly polluted cities. Curcumin has been proven to exert a protective action in the central nervous system (CNS) of diverse experimental models, with no side effects. The aim of this work is to evaluate the effect of curcumin in a preventive and therapeutic manner against the astrocytosis, microgliosis, and apoptosis induced by ozone in rat hippocampus. Fifty Wistar rats were distributed into five experimental groups: The intact control, curcumin fed control, ozone-exposed group, and the preventive and therapeutic groups receiving the curcumin supplementation while exposed to ozone. Ozone caused astrocytosis and microgliosis, as well as apoptosis in the hippocampus. Meanwhile, curcumin was able to decrease the activation of microglia and astrocytes, and apoptotic cell death in both periods of exposure. Therefore, we propose that curcumin could be used as a molecule capable of counteracting the damage caused by ozone in the CNS.
    Keywords:  apoptosis; astrocyte; curcumin; hippocampus; microglia; ozone
    DOI:  https://doi.org/10.3390/molecules24152839
  55. Cell Death Discov. 2019 ;5 124
      Calcium crystal internalization into proximal tubular (PT) cells results in acute kidney injury, nephrocalcinosis, chronic kidney disease (CKD), and kidney-stone formation. Ca2+ supersaturation in PT luminal fluid induces calcium crystal formation, leading to aberrant crystal internalization into PT cells. While such crystal internalization produces reactive oxygen species (ROS), cell membrane damage, and apoptosis; the upstream signaling events involving dysregulation of intracellular Ca2+ homeostasis and ER stress, remain largely unknown. We have recently described a transepithelial Ca2+ transport pathway regulated by receptor-operated Ca2+ entry (ROCE) in PT cells. Therefore, we examined the pathophysiological consequence of internalization of stone-forming calcium crystals such as calcium phosphate (CaP), calcium oxalate (CaOx), and CaP + CaOx (mixed) crystals on the regulation of intracellular Ca2+ signaling by measuring dynamic changes in Ca2+ transients in HK2, human PT cells, using pharmacological and siRNA inhibitors. The subsequent effect on ER stress was measured by changes in ER morphology, ER stress-related gene expression, endogenous ROS production, apoptosis, and necrosis. Interestingly, our data show that crystal internalization induced G-protein-coupled receptor-mediated sustained rise in intracellular Ca2+ concentration ([Ca2+]i) via store-operated Ca2+ entry (SOCE); suggesting that the mode of Ca2+ entry switches from ROCE to SOCE following crystal internalization. We found that SOCE components-stromal interacting molecules 1 and 2 (STIM1, STIM2) and ORAI3 (SOCE) channel were upregulated in these crystal-internalized cells, which induced ER stress, ROS production, and cell death. Finally, silencing those SOCE genes protected crystal-internalized cells from prolonged [Ca2+]i rise and ER stress. Our data provide insight into the molecular mechanism of crystal-induced Ca2+ dysregulation, ER stress, and PT cell death and thus could have a translational role in treating crystal nephropathies including kidney stones. Taken together, modulation of Ca2+ signaling can be used as a tool to reverse the pathological consequence of crystal-induced conditions including cardiovascular calcification.
    Keywords:  Calcium signalling; Mechanisms of disease; Stress signalling
    DOI:  https://doi.org/10.1038/s41420-019-0203-5
  56. Int J Biol Macromol. 2019 Aug 01. pii: S0141-8130(19)32878-8. [Epub ahead of print]
      Cisplatin (CP), a common chemotherapy drug used in treatment of malignant tumors. Due to various side effects such as nephrotoxicity (kidney damage), it's efficiency and therapeutic application are limited. This study focuses on finding a suitable drug that would attenuate the side effects like kidney damage, caused by CP. Huaier polysaccharide (HP-1), an extraction of Trametes robiniophila Murr, with a molecular weight of 3 × 104 Da. Previous studies have shown that HP-1, exhibits anti-tumor potential and immunomodulatory effects. We hypothesized that HP-1 has the effect of attenuating the nephrotoxicity caused by CP chemotherapy and protecting renal function. Through our experiments, we observed that HP-1 can attenuate the level of oxidative stress, inflammation and mitochondrial dysfunction, thereby reducing kidney damage. In vitro, we observed that HP-1 significantly inhibits CP-induced renal tubular cell apoptosis and cell cycle arrest. In addition, HP-1 also affects the expression level of the protein by regulating the PI3K/Akt/mTOR signaling pathway and thus attenuates the side effects induced by cisplatin. Therefore, HP-1 may be a potential drug for preventing CP-induced renal damage.
    Keywords:  Cisplatin; Huaier polysaccharide (HP-1); Kidney damage
    DOI:  https://doi.org/10.1016/j.ijbiomac.2019.07.219
  57. J Photochem Photobiol B. 2019 Jul 25. pii: S1011-1344(19)30615-3. [Epub ahead of print]198 111564
      Light irradiation has been used in clinical therapy for several decades. In this context, photobiomodulation (PBM) modulates signaling pathways via ROS, ATP, Ca2+, while photodynamic therapy (PDT) generates reactive oxygen species by excitation of a photosensitizer. NO generation could be an important tool when combined with both kinds of light therapy. By using a metal-based compound, we found that PBM combined with PDT could be a beneficial cancer treatment option. We used two types of ruthenium compounds, ([Ru(Pc)], Pc = phthalocyanine) and trans-[Ru(NO)(NO2)(Pc)]. The UV-vis spectra of both complexes displayed a band in the 660 nm region. In the case of 0.5 μM trans-[Ru(NO)(NO2)(Pc)], light irradiation at the Q-band reduced the percentage of viable human melanoma (A375) cells to around 50% as compared to [Ru(Pc)]. We hypothesized that these results were due to a synergistic effect between singlet oxygen and nitric oxide. Similar experiments performed with PDT (660 nm) combined with PBM (850 nm) induced more photocytotoxicity using both [Ru(Pc)] and trans-[Ru(NO)(NO2)(Pc)]. This was interpreted as PBM increasing cell metabolism (ATP production) and the consequent higher uptake of the ruthenium phthalocyanine compounds and more efficient apoptosis. The use of metal-based photosensitizers combined with light therapy may represent an advance in the field of photodynamic therapy.
    Keywords:  Light irradiation therapy; Metal-based drug; Nitrosyl ruthenium-phthalocyanine complex; Photobiomodulation
    DOI:  https://doi.org/10.1016/j.jphotobiol.2019.111564
  58. Chem Biol Interact. 2019 Jul 31. pii: S0009-2797(19)30720-3. [Epub ahead of print]311 108777
      Nicorandil ameliorated doxorubicin-induced nephrotoxicity; this study aimed to show and explain the mechanism of this protection. A precise method was elucidated to study the effect of nicorandil on doxorubicin-induced nephrotoxicity in rats depending on the critical inflammation pathway TLR4/MAPK P38/NFκ-B. Adult male rats were subdivided into four groups. The 1st group was normal control, the 2nd group received nicorandil (3 mg/kg; p.o., for 4 weeks), the 3rd group received doxorubicin (2.6 mg/kg, i.p., twice per week for 4 weeks), and the fourth group was combination of doxorubicin and nicorandil for 4 weeks. Nephrotoxicity was assessed by biochemical tests through measuring Kidney function biomarkers such as [serum levels of urea, creatinine, albumin and total protein] besides renal kidney injury molecule-1 (KIM-1) and cystatin C], oxidative stress parameters such as [renal tissue malondialdehyde (MDA), reduced glutathione (GSH), SOD, catalase and nrf-2], mediators of inflammation such as [Toll like receptor 4 (TLR-4), Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), p38 MAPK, Interleukin 1 beta (IL-1 β), and Tumor necrosis factor alpha (TNF-α)] and markers of apoptosis [BAX and Bcl-2 in renal tissue]. Finally, our data were supported by histopathology examination. Nicorandil pretreatment resulted in a significant decrease in nephrotoxicity biomarkers, oxidative stress markers, inflammatory mediators and prevented apoptosis through decreasing BAX and increasing Bcl-2 in renal tissues. Nicorandil prevented all the histological alterations caused by doxorubicin. Nicorandil is a promising antidote against doxorubicin-induced nephrotoxicity by neutralizing all toxicity mechanisms caused by doxorubicin through normalizing inflammatory cascade of TLR4/MAPK P38/NFκ-B.
    Keywords:  Apoptosis; Doxorubicin; Inflammation; Nephrotoxicity; Nicorandil
    DOI:  https://doi.org/10.1016/j.cbi.2019.108777
  59. Pharmacol Rep. 2019 Apr 09. pii: S1734-1140(18)30734-5. [Epub ahead of print]71(5): 774-781
      BACKGROUND: Pirfenidone (PFD) is an orally active antifibrotic agent that has anti-inflammatory activity in diverse animal models. Its effect against acute pancreatitis (AP) has not been elucidated. Hence, the present investigation was carried out to assess the potential protective role of PFD against l-arginine-induced AP in mice.METHODS: AP was induced in adult male Swiss albino mice via intraperitoneal injections of l-arginine (4 g/kg, twice each 1 h apart). PFD (250 mg/kg, orally) was administered one day before and on the day of l-arginine challenge. Twenty-four hours after l-arginine injection, the severity of AP was evaluated using biochemical and histological analyses. Indices of oxidative stress, inflammation and apoptosis were evaluated using ELISA and immunohistochemistry (IHC).
    RESULTS: PFD suppressed the development of l-arginine-induced AP as revealed by the improvement of histopathological lesions of pancreatic specimen and the significant reduction of serum amylase and lipase levels. Notably, PFD reduced the lipid peroxidation and enhanced the antioxidants such as reduced glutathione (GSH) and superoxide dismutase (SOD) in pancreatic tissue. Importantly, PFD suppressed AP-associated elevation of inflammatory cytokines along with depression of nuclear factor kappa-B (NF-κB) immuno-expression in pancreatic tissue. Lastly, PFD efficiently ameliorated AP-induced elevation of the pro-apoptotic protein (Bax) and increased AP-induced reduction of the anti-apoptotic protein (Bcl2).
    CONCLUSIONS: PFD protected against l-arginine-induced AP in mice through anti-oxidative, anti-inflammatory and anti-apoptotic properties.
    Keywords:  Apoptosis; Inflammation; NF-κB; Pancreatitis; Pirfenidone
    DOI:  https://doi.org/10.1016/j.pharep.2019.04.005
  60. Med Sci Monit. 2019 Aug 03. 25 5776-5784
      BACKGROUND The purpose of this study was to investigate the role and mechanism of steroid receptor coactivator-interacting protein (SIP) in an astrocyte model of 1-methyl-4-phenylpyridinium (MPP⁺)-induced Parkinson's disease. MATERIAL AND METHODS To perform our study, a Parkinson's disease cell model was established by treating the rat glioblastoma cell line C6 with MPP⁺. SIP was overexpressed in C6 cells using SIP-plasmid. Cell viability and apoptosis were analyzed using MTT assay and flow cytometer respectively. Tumor necrosis factor (TNF)-alpha and interleukin (IL)-1ß levels were detected using enzyme linked immunosorbent assay and quantitative reverse transcription PCR. Besides, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) production, and superoxide dismutase (SOD) enzyme activity were determined in the present study. For protein and mRNA detection, western blot assay, and qRT-PCR were performed respectively. RESULTS SIP was decreased in MPP⁺-induced C6 cells. SIP overexpression relieved MPP⁺-induced cytotoxicity of C6 cells, displayed as increased cell viability and reduced cell apoptosis and reduced LDH release. Besides, SIP inhibited MPP⁺-induced inflammatory response and oxidative stress, evidenced by decreased levels of inflammatory factors (TNF-alpha and IL-1ß), reduced ROS generation and enhanced SOD activity. Moreover, MPP⁺-induced nuclear factor-kappaB activation was inhibited by SIP overexpression. CONCLUSIONS SIP was downregulated in Parkinson's disease and it played a protective role in the development Parkinson's disease, thus may be a promising target for Parkinson's disease treatment.
    DOI:  https://doi.org/10.12659/MSM.912106
  61. Transplant Proc. 2019 Jul - Aug;51(6):pii: S0041-1345(18)31439-8. [Epub ahead of print]51(6): 2051-2059
      PURPOSE: Hepatic ischemia-reperfusion (IR) injury is a serious complication of many clinical conditions, which may lead to liver or multiple organ failure. Hyperoside, a flavonoid compound, has been reported to protect against myocardial and cerebral injury induced by IR. This study aimed to investigate the protective effects of hyperoside on hepatic IR injury in rats.METHODS: Using the 70% hepatic IR injury model, we divided 32 male Wistar rats into 4 groups (n = 8): sham-operated, IR+saline (saline/p.o.), IR+vehicle (carboxy methyl cellulose/p.o.), and IR+hyperoside (50 mg/kg/d/p.o.). At 24 hours after reperfusion, blood and liver tissue were collected. The effects of hyperoside on hepatic IR injury were assessed through tests of serum transaminase, hepatic histopathology, and measurement of markers of oxidative stress and apoptosis.
    RESULTS: Pretreatment with hyperoside protected the liver from IR injury by a reduction in serum aspartate aminotransferase/alanine aminotransferase levels and a decrease in the severity of histologic changes. Hyperoside treatment also decreased the activity of malondialdehyde, increased the activities of superoxide dismutase and glutathione peroxidase, up-regulated the expression of heme oxygenase 1 and NAD(P)H:quinone oxidoreductase 1, and reduced the apoptotic index after IR injury. A decrease in the expression of caspase-3 and an increase in the ratio of B cell lymphoma 2 to B cell lymphoma 2-associated X also were observed.
    CONCLUSION: Hyperoside has a protective effect on hepatic IR injury in rats, which may be due to its antioxidant and antiapoptotic properties.
    DOI:  https://doi.org/10.1016/j.transproceed.2019.04.066
  62. Biochim Biophys Acta Gen Subj. 2019 Aug 03. pii: S0304-4165(19)30194-1. [Epub ahead of print]
      BACKGROUND: Xanthohumol (XN, a hop-derived prenylflavonoid) was found to exert anticancer effects on various cancer types. However, the mechanisms by which XN affects the survival of multiple myeloma cells (MM) are little known. Therefore, our study was undertaken to address this issue.METHODS: Anti-proliferative activity of XN towards two phenotypically distinct MM cell lines U266 and RPMI8226 was evaluated with the MTT and BrdU assays. Cytotoxicity was determined with the LDH method, whereas apoptosis was assessed by flow cytometry and fluorescence staining. The expression of cell cycle- and apoptosis-related proteins and the activation status of signaling pathways were estimated by immunoblotting and ELISA assays.
    RESULTS: XN reduced the viability of RPMI8226 cells more potently than in U266 cells. It blocked cell cycle progression through downregulation of cyclin D1 and increased p21 expression. The marked apoptosis induction in the XN-treated RPMI8226 cells was related to initiation of mitochondrial and extrinsic pathways, as indicated by the altered p53, Bax, and Bcl-2 protein expression, cleavage of procaspase 8 and 9, and elevated caspase-3 activity. The apoptotic process was probably mediated via ROS overproduction and MAPK (ERK and JNK) activation as N-acetylcysteine, or specific inhibitors of these kinases prevented the XN-induced caspase-3 activity and, hence, apoptosis. Moreover, XN decreased sIL-6R and VEGF production in the studied cells.
    CONCLUSIONS: ERK and JNK signaling pathways are involved in XN-induced cytotoxicity against MM cells.
    GENERAL SIGNIFICANCE: The advanced understanding of the molecular mechanisms of XN action can be useful in developing therapeutic strategies to treat multiple myeloma.
    Keywords:  Apoptosis; MAPKs; Multiple myeloma; ROS; VEGF; Xanthohumol; sIL-6R
    DOI:  https://doi.org/10.1016/j.bbagen.2019.08.001
  63. Adv Healthc Mater. 2019 Aug 07. e1900612
      In this study, hybrid nanocubes composed of magnetite (Fe3 O4 ) and manganese dioxide (MnO2 ), coated with U-251 MG cell-derived membranes (CM-NCubes) are synthesized. The CM-NCubes demonstrate a concentration-dependent oxygen generation (up to 15%), and, for the first time in the literature, an intracellular increase of temperature (6 °C) due to the exothermic scavenging reaction of hydrogen peroxide (H2 O2 ) is showed. Internalization studies demonstrate that the CM-NCubes are internalized much faster and at a higher extent by the homotypic U-251 MG cell line compared to other cerebral cell lines. The ability of the CM-NCubes to cross an in vitro model of the blood-brain barrier is also assessed. The CM-NCubes show the ability to respond to a static magnet and to accumulate in cells even under flowing conditions. Moreover, it is demonstrated that 500 µg mL-1 of sorafenib-loaded or unloaded CM-NCubes are able to induce cell death by apoptosis in U-251 MG spheroids that are used as a tumor model, after their exposure to an alternating magnetic field (AMF). Finally, it is shown that the combination of sorafenib and AMF induces a higher enzymatic activity of caspase 3 and caspase 9, probably due to an increment in reactive oxygen species by means of hyperthermia.
    Keywords:  cell membrane-coated nanocubes; glioblastoma; homotypic targeting; magnetic hyperthermia; theranostics
    DOI:  https://doi.org/10.1002/adhm.201900612
  64. Front Pharmacol. 2019 ;10 802
      Objective: FTY720, an immunomodulator derived from sphingosine-1-phosphate, has recently demonstrated its immunomodulatory, anti-inflammatory, anti-oxidant, anti-apoptotic and anti-inflammatory properties. Furthermore, FTY720 might be a key pharmacological target for preconditioning. In this preclinical model, we have investigated the effects of FTY720 on myocardium during reperfusion in an experimental model of cardioplegic arrest (CPA) and cardiopulmonary bypass. Methods: 30 Sprague-Dawley rats (300-350 g) were randomized into two groups: Group-A, treated with FTY720 1 mg/kg via intravenous cannulation, and Group-B, as control. After 15 min of treatment, rats underwent CPA for 30 min followed by initiation of extracorporeal life support for 2 h. Support weaning was done, and blood and myocardial tissues were collected for analysis. Hemodynamic parameters, inflammatory mediators, nitro-oxidative stress, neutrophil infiltration, immunoblotting analysis, and immunohistochemical staining were analyzed and compared between groups. Results: FTY720 treatment activated the Akt/Erk1/2 signaling pathways, reduced the level of inflammatory mediators, activated antiapoptotic proteins, and inhibited proapoptotic proteins, leading to reduced nitro-oxidative stress and cardiomyocyte apoptosis. Moreover, significant preservation of high-energy phosphates were observed in the FTY720-treated group. This resulted in improved recovery of left ventricular systolic and diastolic functions. Conclusion: The cardioprotective mechanism in CPA is associated with activation of prosurvival cell signaling pathways that prevents myocardial damage. FTY720 preserves high-energy phosphates attenuates myocardial inflammation and oxidative stress, and improves cardiac function.
    Keywords:  FTY 720; anti-inflammatory; cardioplegic arrest; cardioprotection; ischemia reperfusion injury
    DOI:  https://doi.org/10.3389/fphar.2019.00802
  65. Cell Mol Life Sci. 2019 Aug 07.
      The majority of anticancer drugs are DNA-damaging agents, and whether or not they may directly target mitochondria remains unclear. In addition, tumors such as neuroblastoma exhibit addiction to glutamine in spite of it being a nonessential amino acid. Our aim was to evaluate the direct effect of widely used anticancer drugs on mitochondrial activity in combination with glutamine withdrawal, and possible apoptotic effects of such interaction. Our results revealed that etoposide inhibits mitochondrial respiratory chain Complex I causing the leakage of electrons and the superoxide radical formation. However, it was not sufficient to induce apoptosis, and apoptotic manifestation was detectable only alongside the withdrawal of glutamine, a precursor for antioxidant glutathione. Thus, the simultaneous depletion of glutathione and destabilization of mitochondria by ROS can compromise the barrier properties of the mitochondrial membrane, leading to cytochrome c release and the activation of the mitochondrial apoptotic pathway. Thus, the depletion of antioxidants or the inhibition of the pathways responsible for cellular antioxidant response can enhance mitochondrial targeting and strengthen antitumor therapy.
    Keywords:  Apoptosis; Mitochondria; Neuroblastoma; Oxidative stress; Respiratory chain
    DOI:  https://doi.org/10.1007/s00018-019-03232-z
  66. Am J Transl Res. 2019 ;11(7): 4470-4480
      Resistance to Gemcitabine (GEM) is a crucial problem in treatment of pancreatic cancer. Many studies indicate the direct impact of glycolytic enzyme on chemoresistance. However, it still has not been known whether Enolase 1 (ENO1), a multifunctional glycolytic enzyme, is a potential target to overcome GEM resistance in pancreatic ductal adenocarcinoma (PDAC). In this study, we showed that ENO1 high expression was associated with poor prognosis of PDAC patients. Moreover, we investigated the impacts of ENO1 silencing on hypoxia induced GEM chemoresistance in CFPAC-1 and MiaPaCa-2 cells. The results showed that, targeting ENO1 using ENO1-shRNA could sensitize hypoxia induced chemoresistance in pancreatic cancer cells by modulation of redox homeostasis, the mechanisms appear to be associated with influences on proliferation, apoptosis, and cell cycle regulated by increased intracellular reactive oxygen species (ROS). We demonstrated that targeting ENO1 could be a potential strategy for overcoming hypoxia induced GEM chemoresistance in PDAC cells.
    Keywords:  Phosphopyruvate hydratase; chemoresistance; gemcitabine; pancreatic neoplasms; redox homeostasis
  67. Int J Angiol. 2019 Jun;28(2): 71-79
      Pulmonary hypertension (PH) is a rare and fatal disease characterized by elevation of pulmonary artery pressure ≥ 25 mm Hg. There are five groups of PH: (1) pulmonary artery (PA) hypertension (PAH), (2) PH due to heart diseases, (3) PH associated with lung diseases/hypoxia, (4) PH associated with chronic obstruction of PA, and (5) PH due to unclear and/or multifactorial mechanisms. The pathophysiologic mechanisms of group 1 have been studied in detail; however, those for groups 2 to 5 are not that well known. PH pathology is characterized by smooth muscle cells (SMC) proliferation, muscularization of peripheral PA, accumulation of extracellular matrix (ECM), plexiform lesions, thromboembolism, and recanalization of thrombi. Advanced glycation end products (AGE) and its receptor (RAGE) and soluble RAGE (sRAGE) appear to be involved in the pathogenesis of PH. AGE and its interaction with RAGE induce vascular hypertrophy through proliferation of vascular SMC, accumulation of ECM, and suppression of apoptosis. Reactive oxygen species (ROS) generated by interaction of AGE and RAGE modulates SMC proliferation, attenuate apoptosis, and constricts PA. Increased stiffness in the artery due to vascular hypertrophy, and vasoconstriction due to ROS resulted in PH. The data also suggest that reduction in consumption and formation of AGE, suppression of RAGE expression, blockage of RAGE ligand binding, elevation of sRAGE levels, and antioxidants may be novel therapeutic targets for prevention, regression, and slowing of progression of PH. In conclusion, AGE-RAGE stress may be involved in the pathogenesis of PH and the therapeutic targets should be the AGE-RAGE axis.
    Keywords:  advanced glycation end products; cell receptor for AGE; pathogenesis; pulmonary hypertension; soluble RAGE; therapeutic targets
    DOI:  https://doi.org/10.1055/s-0039-1687818
  68. BMC Complement Altern Med. 2019 Aug 07. 19(1): 204
      BACKGROUND: Metastasis is the main cause of lung cancer death. As a seed of metastasis, circulating tumor cells are an important target for metastasis intervention. The traditional Chinese medicine, Jinfukang, has been clinically available for the treatment of non-small cell lung cancer (NSCLC). In this study, we investigated the action and underlying mechanisms of Jinfukang against circulating lung tumor cells.METHODS: The cell counting kit-8 (CCK-8), colony formation and cell cycle assays were used to study the cell proliferation ability. Flow cytometry was used to detect the apoptosis and the expression level of ROS and Caspase-3. Comet and TUNEL assays were used to detect DNA damage. DNA damage related pathway protein was detected by western blot.
    RESULTS: Jinfukang significantly inhibits the proliferation of CTC-TJH-01 cells by inducing G1 phase arrest and inhibits their colony formation in a dose-dependent manner. Moreover, Jinfukang induces apoptosis in CTC-TJH-01 cells through the ROS-mediated ATM/ATR-p53 pathway and DNA damage.
    CONCLUSIONS: Our findings suggest that Jinfukang may be a potential drug for lung cancer metastasis.
    Keywords:  Apoptosis; Circulating tumor cell; Jinfukang; Non-small cell lung cancer; Oxidative stress
    DOI:  https://doi.org/10.1186/s12906-019-2601-x
  69. Methods Mol Biol. 2019 Aug 08.
      Photodynamic therapy is a promising, minimally invasive, and clinically approved treatment strategy that destroys the cell components by oxidizing the biological molecules such as nucleic acids, carbohydrates, proteins, and lipids, and leads apoptosis in the cells of the target tissue through the generation of singlet oxygen and reactive oxygen species (ROS) owing to the synergic interactions of a nontoxic photosensitizer, a non-thermal light source, and tissue oxygen. This innovative method has drawn the attention of many scientists and been employed in a wide range of medical fields that covers the treatment of cancer diseases and precancerous dermatological disorders, and the aesthetic and cosmetic practices, including photorejuvenation and treatment of photoaging, hirsutism, facial flat warts, rosacea, acne vulgaris, and sebaceous gland hyperplasia. It was therefore intended to provide an in vitro photodynamic therapy assay protocol on human healthy keratinocytes and epidermoid carcinomas to investigate comparatively the therapeutic and destructive activities of the potent light-sensitive medications.
    Keywords:  Cutaneous squamous cell carcinoma; Fluence; Fluence rate; Human epidermoid carcinoma; Human keratinocyte; Irradiation; Photodynamic therapy; Photosensitizer; Skin cancer; Visible light
    DOI:  https://doi.org/10.1007/7651_2019_260
  70. J Virol. 2019 Aug 07. pii: JVI.00653-19. [Epub ahead of print]
      Equine infectious anemia virus (EIAV) is an equine lentivirus similar to HIV-1, targets to host immune cells and causes life-long infection in horses. The Chinese live EIAV vaccine is attenuated from long-term passaging of a high virulent strain in vitro The parent pathogenic strain (EIAVDLV34) induces a host inflammatory storm to cause severe pathological injury of animals. However, the vaccine strain (EIAVDLV121) induces a high level of apoptosis to eliminate the infected cells. To investigate how these processes are regulated, we performed a comparative proteomics analysis and functional study in equine monocyte-derived macrophages (eMDMs), and found that divergent mitochondrial protein expression profiles caused by EIAV strains with different virulence lead to disparate mitochondrial function, morphology and metabolism. This in turn promoted distinct transformation of macrophage inflammatory polarization and intrinsic apoptosis. In EIAVDLV34 infected cells, a high level of glycolysis and increased mitochondrial fragmentation were induced, resulting in M1-polarized pro-inflammatory type transformation of macrophages and subsequently producing a strong inflammatory response. Following infection with EIAVDLV121, the infected cells were transformed into M2-polarized anti-inflammatory macrophages by inhibition of glycolysis. In this case, decrease of mitochondrial membrane potential and impairment of electronic respiratory chain led to increased levels of apoptosis and ROS. These results are correlated with the viral pathogenicity loss and may help to understand the key mechanism of lentiviral attenuation.IMPORTANCEFollowing viral infection, the working pattern and function of the cell can be transformed through the impact on mitochondria. It still unknown how mitochondrial response changes in the cells infected with viruses in the process of virulence attenuation. EIAVDLV121 is the only effective lentiviral vaccine for large-scale use in world. EIAVDLV34 is a parent pathogenic strain. Unlike EIAVDLV34-induced inflammation storms, EIAVDLV121 can induce high levels of apoptosis. For the first time, we found that, after altering mitochondrial protein expression profile, EIAVDLV34 infected cells are transformed into M1-polarized type macrophages to cause inflammatory injury and the intrinsic apoptosis pathway is activated in EIAVDLV121 infected cells. These studies shed light on how the mitochondrial protein expression profile change from cells infected by pathogenic or attenuated lentivirus strains to drive different cellular response, especially from inflammation to apoptosis.
    DOI:  https://doi.org/10.1128/JVI.00653-19
  71. Adv Pharm Bull. 2019 Jun;9(2): 302-306
      Purpose: Aim of present work was to study cytoprotective properties of lithium pyruvate, as a prospective pharmacological agent. Pyruvate has a lot of potential benefits due to positive influence on cell metabolism. Lithium is "gold-standard" mood-stabilizer. Combination of both may lead advantages. Methods: Lithium pyruvate was tested as cytoprotector on human blood mononuclears under induced oxidative stress. Cells were obtained from healthy donors and patients with alcoholism. The detection of cell viability, apoptosis and determination of oxidative stress level were conducted by flow cytometry. Results: Lithium pyruvate showed excellent cytoprotective properties in normal and oxidation conditions. This effect was independent from cell donor health status. It was shown on cells from healthy donors and alcoholics patients. Conclusion: Obtained results allow considering lithium pyruvate as potential normothymic agents (mood stabilizer) with excellent cytoprotective properties.
    Keywords:  Antioxidant; Apoptosis; Cytoprotection; Lithium pyruvate; Mood-stabilizer; Oxidative stress
    DOI:  https://doi.org/10.15171/apb.2019.035
  72. Environ Sci Pollut Res Int. 2019 Aug 07.
      The purpose of this study was therefore to investigate the effects of radiotherapy on the kidney and the potential use of agents such as N-acetylcysteine (NAC) in developing a future therapeutic protocol for radiation-induced nephrotoxicity at the histopathological and biochemical levels. Our study consisted of three groups: control (oral saline solution only; group 1), irradiation (IR; group 2), and NAC + IR (group 3). The irradiation groups received a single dose of whole-body 6-Gy x-irradiation. The NAC group received 300 mg/kg by the oral route for 7 days, from 5 days before irradiation to 2 days after. All subjects were sacrificed under anesthesia 2 days after irradiation. IR increased tubular necrosis scores (TNS), MDA, and caspase-3 expression, while reducing renal tissue GSH levels. We also observed dilation in renal corpuscles and tubules. Capillary congestion was present in the intertubular spaces. NAC reduced the levels of TNS, MDA, and caspase-3 expression, but increased the levels of renal tissue GSH. ROS-scavenging antioxidants may represent a promising means of preventing renal injury in patients undergoing radiotherapy.
    Keywords:  Apoptosis; Kidney; N-Acetylcysteine; Oxidative stress; Radiation; Rat
    DOI:  https://doi.org/10.1007/s11356-019-06110-0
  73. Peptides. 2019 Aug 03. pii: S0196-9781(19)30099-3. [Epub ahead of print] 170121
      Endothelial dysfunction is considered to be an initial indicator in diabetes-induced macrovascular complications. Evidence has shown that CGRP is an important neuropeptide active in vascular system, especially in vasorelaxation. This study aimed to investigate the role of CGRP in high-glucose-induced endothelial dysfunction in rat aorta endothelial cells (RAECs). Quantitative-real time PCR and western blots were used to determine the efficiency of overexpression and interference of CGRP. After incubation with normal glucose (5.5 mM) or high glucose (33 mM), the cell viability and cell apoptosis were tested. Afterwards, the Nitric Oxide (NO) production, the mRNA expression of inducible nitric oxide synthase (iNOS), endothelial nitric oxide synthase (eNOS) and angiotensin II (Ang II) and the level of reactive oxygen species (ROS) were determined. The involvement of ERK1/2-NOX4 was determined through western blots and the translocation of p47phox was also observed via cell immunofluorescence. CGRP alleviated the high-glucose-induced cell apoptosis while CGRP did not have an obvious impact on cell viability. Meanwhile, CGRP increased the NO production as well as the eNOS mRNA expression and reversely decreased the stimulated expression of iNOS and Ang II by high glucose. In addition, CGRP attenuated the high-glucose-stimulated intracellular ROS production by ERK1/2-NOX4 and the translocation of p47phox. These results indicated the protective role of CGRP in high-glucose-induced oxidative injury in RAECs possibly through inhibiting ERK1/2-NOX4. Our findings might help to further understand the potential role and possible mechanism of CGRP in endothelial dysfunction caused by high glucose.
    Keywords:  Calcitonin gene-related peptide; Diabetes mellitus; Nitric Oxide; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.peptides.2019.170121