bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2018‒12‒16
78 papers selected by
Gavin McStay
Staffordshire University


  1. Oxid Med Cell Longev. 2018 ;2018 7426950
      Camalexin is a phytoalexin that accumulates in various cruciferous plants upon exposure to environmental stress and plant pathogens. It was shown that camalexin has potent antitumor properties, but its underlying mechanisms are still elusive. In the present study, we evaluated the effects of camalexin on human leukemic cells and normal polymorph nuclear cells. CCK-8 assay was used to determine cell viability after camalexin treatment. Apoptosis, intracellular reactive oxygen species (ROS) levels, and loss of mitochondrial membrane potential (MMP) were measured by flow cytometry. The activity of SOD, catalase, and ratio of GSH/GSSG were assayed. ER stress and apoptotic signaling pathway was examined by Western blot. Xenograft mice were used to verify the effect of camalexin in vivo. Our results indicated that camalexin inhibited viability of leukemic but not normal polymorph nuclear cells. Furthermore, camalexin induces apoptosis via the mitochondrial pathway in a caspase-dependent manner. We also observed ER stress is located upstream of apoptosis induced by camalexin. Besides, ROS levels, SOD activity, CAT activity, and GSSG levels were significantly enhanced while the GSH level was decreased after treatment of camalexin. In addition, the generation of ROS is critical for the ER stress and apoptosis induced by camalexin. Finally, administration of camalexin suppresses xenograft tumor graft growth without obvious toxicity. Taken together, this study indicates that camalexin exerts antitumor effects against leukemia cells via the ROS-ER stress-mitochondrial apoptosis pathway.
    DOI:  https://doi.org/10.1155/2018/7426950
  2. Nanomaterials (Basel). 2018 Dec 12. pii: E1040. [Epub ahead of print]8(12):
      Anticancer metallodrugs that aim to physiological characters unique to tumor microenvironment are expected to combat drug tolerance and side-effects. Recently, owing to the fact that reactive oxygen species' is closely related to the development of tumors, people are committed to developing metallodrugs with the capacity of improving the level of reactive oxygen species level toinduce oxidative stress in cancer cells. Herein, we demonstrated that peptide templated gold clusters with atomic precision preferably catalyze the transformation of hydrogen peroxide into superoxide anion in oxidative pressure-type tumor cells. Firstly, we successfully constructed gold clusters by rationally designing peptide sequences which targets integrin ανβ₃ overexpressed on glioblastoma cells. The superoxide anion, radical derived from hydrogen peroxide and catalyzed by gold clusters, was confirmed in vitro under pseudo-physiological conditions. Then, kinetic parameters were evaluated to verify the catalytic properties of gold clusters. Furthermore, these peptide decorated clusters can serve as special enzyme-like catalyst to convert endogenous hydrogen peroxide into superoxide anion, elevated intracellular reactive oxygen species levels, lower mitochondrial membrane potential, damage biomacromolecules, and trigger tumor cell apoptosis consequently.
    Keywords:  biocatalysis; gold clusters; oxidative pressure-type tumor; reactive oxygen species
    DOI:  https://doi.org/10.3390/nano8121040
  3. Oncol Rep. 2018 Oct 31.
      Cinobufagin is a cardiotoxic bufanolide steroid secreted by the Asiatic toad Bufo gargarizans. Cinobufagin is one of the active ingredients in the anticancer Chinese medicine called Chan Su, which was demonstrated to be an effective treatment for gastric cancer. Increasing evidence shows that inhibition of autophagy has a pro‑apoptotic effect on human gastric cancer cells. The aim of the present study was to investigate the relationship between cinobufagin, autophagy and apoptosis in gastric cancer. Autophagy was induced or inhibited in the human gastric cancer cell line SGC‑7901 by incubation in HBSS media or by treatment with 3‑methyladenine or ATG5 siRNA, respectively. Following treatment, the levels of apoptosis, apoptotic proteins, reactive oxygen species (ROS), and mitochondrial membrane potential were compared between the conditions. As anticipated, we found that cinobufagin increased apoptosis in SGC‑7901 cells. Notably, inhibition of autophagy, monitored by the absence of the autophagosome marker LC3‑II, also enhanced cell apoptosis. This effect was reversed when autophagy was induced by incubation in HBSS media. Enhanced expression of pro‑apoptotic indicators, including BAX, cytosolic cytochrome c, cleaved PARP, caspase‑3 and caspase‑9, was detected when autophagy was suppressed. Increased pro‑apoptotic protein expression was accompanied by disrupted mitochondrial membrane potential and elevated ROS production. Altogether, these data suggest that inhibition of autophagy enhances the anticancer action of cinobufagin through increased apoptosis of gastric cancer cells. Moreover, these effects may be partly mediated by ROS generation and the activation of the mitochondrial programmed cell death pathway.
    DOI:  https://doi.org/10.3892/or.2018.6837
  4. Biomed Pharmacother. 2018 Dec 08. pii: S0753-3322(18)33691-6. [Epub ahead of print]110 602-608
      BACKGROUND: Osteoblasts play important roles in the process of osteogenesis and prevention of osteonecrosis. Dexamethasone (Dex), a type of glucocorticoids (GCs), induces apoptosis of osteoblasts and leads to the occurrence of non-traumatic osteonecrosis. This study aimed to explore the effects of phosphatidylinositol 3-kinase/Protein kinase 3 (PI3K/AKT) and glycogen synthase kinase 3β (GSK3β) on Dex-induced osteoblasts apoptosis.METHODS: Viabilities, proliferation, and apoptosis of primary osteoblasts and pre-osteoblast MC3T3-E1 cells after Dex treatment were detected using cell counting kit-8 (CCK-8) assay, 5-bromo-2'-deoxyuridine (BrdU) incorporation assay, FITC-Annexin V/PI staining and western blotting, respectively. 2',7'-Dichlorodihydrofluorescein diacetate (DCFH-DA) staining was performed to measure the intracellular reactive oxygen species (ROS) levels after Dex treatment. N-acetyl-l-cysteine (NAC) was used as ROS scavenger in this research. The expressions of PI3K/AKT and GSK3β in osteoblasts and MC3T3-E1 cells after Dex treatment were analyzed using western blotting and qRT-PCR, respectively. Then the effects of GSK3β knockdown on Dex-induced apoptosis of osteoblasts were explored. Alkaline phosphatase (ALP) activity assay was used to detect the role of Dex in regulating ALP activity.
    RESULTS: Dex remarkably inhibited proliferation and induced apoptosis of osteoblasts and MC3T3-E1 cells. Dex potentially attenuated the osteoblast differentiation. The intracellular ROS levels were significantly increased after Dex treatment. Dex suppressed the activation of PI3K/AKT pathway in osteoblasts and MC3T3-E1 cells by down-regulating the expressions of p-PI3K and p-AKT. The expressions of GSK3β in osteoblasts and MC3T3-E1 cells were obviously up-regulated after Dex treatment. Knockdown of GSK3β alleviated Dex-induced osteoblast and MC3T3-E1 cell apoptosis by decreasing the expressions of Bax, cleaved-caspase 3, cleaved-caspase 9 and increasing the expression of Bcl-2.
    CONCLUSION: Our research verified that Dex induced osteoblasts apoptosis by ROS-PI3K/AKT/GSK3β signaling pathway.
    Keywords:  Dexamethasone; Glycogen synthase kinase 3β; Osteoblasts apoptosis; Osteonecrosis; Phosphatidylinositol 3-kinase/protein kinase 3; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.biopha.2018.11.103
  5. Int J Mol Med. 2018 Dec 03.
      The present study investigated the mechanisms of apoptosis induced by cryptotanshinone (CT) in human rheumatoid arthritis fibroblast‑like synoviocytes (RA‑FLSs). Cell Counting kit‑8 assay was performed to determine the cytotoxic effects of CT in human RA‑FLSs, including primary RA‑FLS, HFLS‑RA and MH7A cells, and in HFLS cells derived from normal synovial tissue. Annexin V‑FITC/PI staining was used to detect the apoptotic effects of CT in HFLS‑RA and MH7A cells. Flow cytometry was performed to detect the apoptotic and reactive oxygen species (ROS) levels induced by CT in HFLS‑RA cells. Western blotting was used to assess the expression levels of proteins associated with apoptosis and with the mitogen‑activated protein kinase (MAPK), protein kinase B (Akt), and signal transducer and activator of transcription‑3 (STAT3) signaling pathways. The results demonstrated that CT treatment significantly suppressed HFLS‑RA and MH7A cell growth, whereas no clear inhibitory effect was observed in normal HFLS cells. CT exposure downregulated the expression levels of B‑cell lymphoma 2 (Bcl‑2), p‑Akt, p‑extracellular signal‑related kinase and p‑STAT3, while it upregulated the expression levels of Bcl‑2‑associated death promoter (Bad), caspase‑3, poly (ADP‑ribose) polymerase (PARP), p‑p38 and p‑c‑Jun N‑terminal kinase. Following ROS scavenging, the CT‑induced apoptosis and altered expression levels of Bcl‑2, Bad, cleaved caspase‑3 and cleaved PARP were restored. Furthermore, the Akt, MAPK and STAT3 signaling pathways were regulated by intracellular ROS. These results suggest that ROS‑mediated Akt, MAPK and STAT3 signaling pathways serve important roles in the CT‑induced apoptosis of RA‑FLSs.
    DOI:  https://doi.org/10.3892/ijmm.2018.4012
  6. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)33871-X. [Epub ahead of print]109 2309-2317
      Cisplatin, as an effective chemotherapeutic agent, is widely used to treat verious types of cancers. Nephrotoxicity induced by cisplatin seriously limits its clinical application. Icariin, a major and remarkable flavonoid isolated from Epimedium koreanum, has been reported to exert anti-oxidative stress and anti-inflammation actions. The purpose of this study is to explore the protective effect and possible mechanism of icariin on cisplatin-induced nephrotoxicity on HEK-293 cells. In this study, icariin pretreatment for 24 h significantly ameliorated cisplatin-induced oxidative stress by reducing levels of malondialdehyde (MDA) and reactive oxygen species (ROS), while increasing level of glutathione (GSH) in HEK-293 cells. Furthermore, icariin pretreatment reduced NF-κB phosphorylation and nuclear translocation in HEK-293 cells followed by decreased secretion of IL-1β, TNF-α, and iNOS, suggesting a suppression of inflammatory response. Moreover, icariin pretreatment significantly reduced cellular apoptosis via reduced levels of Bax, cleaved caspase-3/9, and increased anti-apoptotic protein Bcl-2 in the cells. Importantly, LY294002, a specific PI3K inhibitor, abrogated the anti-apoptosis effect of icariin, implicating the involvement of PI3K/Akt pathway. In summary, icariin prevents cisplatin-induced HEK-293 cell injury by inhibiting oxidative stress, inflammatory response, and cellular apoptosis partly via regulating NF-κB and PI3K/Akt signaling pathways. Icariin may serve as a potential therapeutic target against cisplatin-induced nephrotoxicity.
    Keywords:  Cisplatin; Cytotoxicity; HEK-293 cells; Icariin; PI3K/Akt
    DOI:  https://doi.org/10.1016/j.biopha.2018.11.108
  7. Int J Biol Macromol. 2018 Dec 03. pii: S0141-8130(18)35597-1. [Epub ahead of print]
      Codonopsis pilosula polysaccharides (CPPS) has been shown to possess a variety of biological activities. In previous study, CPPS was successfully modified to obtain its best selenizing Codonopsis pilosula polysaccharides (sCPPS5). The purpose of this study was to investigate the protective effect of the selenizing derivative of CPPS (sCPPS5) from H2O2-induced oxidative damage in RAW264.7 murine macrophages and the possible mechanism of this protection. Results showed that the sCPPS5 was significantly stronger than that of the corresponding unmodified polysaccharide, CPPS. Meanwhile, sCPPS5 treatment could improve the production of reactive oxygen species (ROS), antioxidant enzyme, MMP, caspases-3 and apoptosis capacity of H2O2-induced RAW264.7 cells. Moreover, the mechanism might be elucidated that sCPPS5 could increase expression level of Nrf2 and its downstream ARE gene battery, promote production of corresponding antioxidative enzymes and protein, and enhance Keap1-Nrf2/ARE signaling pathway to avoid male reproductive dysfunction. Overall, these results showed that sCPPS5 as a potent antioxidant could reduce reproductive oxidative stress damage related to Keap1-Nrf2/ARE pathway.
    Keywords:  Keap1-Nrf2/ARE pathway; Oxidative stress; Selenizing Codonopsis pilosula polysaccharide
    DOI:  https://doi.org/10.1016/j.ijbiomac.2018.12.025
  8. Eur J Pharmacol. 2018 Dec 06. pii: S0014-2999(18)30703-9. [Epub ahead of print]
      Despite the triumph of highly active antiretroviral therapy (HAART) in anti-HIV infection, more than half of the HIV infection individuals receiving antiretroviral therapy acquire HIV-associated neurocognitive disorder (HAND). Previously researches had reported that the HAART neurotoxicity is implicated in HAND-related morbidity. The molecular mechanism of HAND is not clear. Tenofovir disoproxil fumarate (TDF) is a novel nucleotide reverse transcriptase inhibitor (NRTI), which was recommended as first-line therapeutic schedule for free AIDS antiviral drugs. Whether the neurotoxicity of TDF is associated with HAND is not well known. In this study, the cell viability of TDF-treated pheochromocytoma cells (PC-12) line was detected using MTT assay, while apoptosis was evaluated by Hoechst 33342 staining, TUNEL assay, as well as flow cytometry. In addition, the level of reactive oxygen species and BAX protein expression were evaluated using DCFH-DA staining and western blotting. The results showed that the proliferation of PC-12 cells was significantly inhibited by TDF. The morphological assay, TUNEL assay and flow cytometry showed that TDF efficiently triggered apoptosis in PC-12 cells. The reactive oxygen species levels were BAX expression was markedly up-regulated in PC-12 cells after treatment with TDF. These findings indicated that TDF may induce PC-12 cell apoptosis. TDF has neural toxicity effect that is relevant to the cell apoptosis, which may be related to the increasing prevalence of HAND.
    Keywords:  HIV-associated neurocognitive disorder; Pheochromocytoma Cells; Tenofovir Disoproxil Fumarate; apoptosis; neurotoxicity
    DOI:  https://doi.org/10.1016/j.ejphar.2018.12.006
  9. Cell Stress Chaperones. 2018 Dec 13.
      Chondrocyte apoptosis is closely related to the development and progression of osteoarthritis (OA); however, the underlying mechanisms remain enigmatic. Previous studies have confirmed that cell apoptosis is one of the main pathological alterations during oxidative stress, and chondrocyte apoptosis induced by oxidative stress plays an important role in the development of OA. Rat chondrocytes exposed to hydrogen peroxide (H2O2) were used as the experimental oxidative stress model. We assessed cell viability, cell apoptosis, levels of intracellular reactive oxygen species (ROS), nitric oxide (NO) production, gene relative expression level of inducible nitric oxide synthase (iNOS), and expressions of iNOS, PI3K, phospho-Akt, caspase-9, and caspase-3. With the rising of intracellular ROS and increasing iNOS synthesis, producing a large amount of NO in chondrocytes, H2O2 decreased the cell viability and induced cell apoptosis of chondrocytes. Furthermore, the levels of caspase-9 and caspase-3 protein expression were significantly elevated as well as the level of p-Akt protein expression when induced by oxidative stress. These findings suggest that oxidative stress-induced chondrocyte apoptosis occurred via activating both PI3K/Akt and caspase pathways in the early stage in these processes.
    Keywords:  Apoptosis; Caspases; Oxidative stress; PI3K/Akt
    DOI:  https://doi.org/10.1007/s12192-018-0956-4
  10. Chem Biol Interact. 2018 Dec 10. pii: S0009-2797(18)30720-8. [Epub ahead of print]
      Approximately 15% of globally diagnosed breast cancers are designated as triple negative breast cancer (TNBC). In this study, we investigated the effect of the natural compound, Bis(2- ethyl hexyl) 1H-pyrrole-3,4-dicarboxylate (TCCP), purified from Tinospora cordifolia on MDA-MB-231, a TNBC cell line. The pro-apoptotic nature of TCCP on MDA-MB-231 was determined by assessing various apoptotic markers. ROS generation, intracellular calcium, mitochondrial membrane potential (ΔΨm), MPTP, cardiolipin peroxidation and caspase activity were determined fluorometrically. BAX, BCL-2, cytochrome c, caspases, and p53 protein expressions were determined by immunoblotting. Further, the effect of TCCP on DNA and cell death was determined by DNA fragmentation assay, annexin-V staining, and cell cycle analysis. TCCP treatment caused endogenous ROS generation, increase in intracellular calcium and phosphorylation of p53 in a concentration-dependent manner, which was reverted upon pre-treatment with pifithrin-μ. This led to the downstream altered expression of Bcl-2 and Bax proteins, mitochondrial membrane depolarization, MPTP, and cardiolipin peroxidation. TCCP induced cytochrome c release into the cytosol, caspase activation, ultimately resulting in DNA fragmentation. Further, induction of apoptosis and morphological alterations were evident from the phosphatidylserine externalization and increase in sub G1 population. The in vivo Ehrlich ascites tumor (EAT) mouse study revealed the effectiveness of TCCP in reducing the tumor burden and resulted in a ∼2 fold increase in mice survival with minimal hepato-renal toxicity. Overall, TCCP was shown to be efficient in inducing ROS and mitochondrial-mediated apoptosis by restoring p53 activity in MDA-MB-231 cells and also induced EAT cell death in vivo thereby inhibiting tumor proliferation.
    Keywords:  Apoptosis; TCCP; TNBC
    DOI:  https://doi.org/10.1016/j.cbi.2018.12.005
  11. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)34478-0. [Epub ahead of print]109 2109-2118
      OBJECTIVES: Scutellarein is a flavonoid monomer found in traditional Chinese medicine such as Scutellaria barbata. This study aimed to investigate the cytotoxic effect of scutellarein treatment on multiple myeloma (MM) cells.METHODS: circulating B lymphocytes (CBL) isolated from healthy donors' peripheral blood served as control for MM.1R and IM-9 MM cells. CLB and MM cells were treated with various concentrations of scutellarein before their cell viability and apoptosis being evaluated. Nude mice burdened with MM xenograft tumor were intravenously injected with different concentrations of scutellarein, and their tumor burden change were monitored. Apoptosis of MM cells or CBL after scutellarein treatment was assayed by measuring caspase-3, -8 and -9 activities. FADD or APAF1 gene knockdown in MM cells was achieved by lentiviral transfection. Amount of Cytochrome C in cytosol or mitochondria as well as that of Bax and Bcl-2 protein were evaluated by Western blot. Mitochondria-induced apoptosis was assayed by measuring mitochondrial membrane potential change. Production of general reactive oxygen species and mitochondrial superoxide in MM or CBL was detected after scutellarein treatment, which was reduced by MitoTEMPO or apocynin treatment, respectively.
    RESULTS: Scutellarein treatment showed potent cytotoxicity on MM cells but not on viable CBL, and intravenous injection of scutellarein significantly reduced MM xenograft tumor burden in nude mice. Scutellarein treatment in MM cells activated the mitochondrial-mediated intrinsic apoptosis pathway by increasing the production of mitochondrial superoxide, which was reduced to ROS by NADPH, but this effect was weakened in healthy CBL. Co-treatment with scutellarein synergized with bortezomib in inducing apoptosis in MM cells in vitro and in reducing tumor volume in MM xenografted nude mice.
    CONCLUSIONS: Scutellarein induced mitochondrial-mediated intrinsic apoptosis selectively on malignant cells comparing to healthy cells.
    Keywords:  Mitochondria; Multiple myeloma; NADPH; ROS; Scutellarein; Superoxide
    DOI:  https://doi.org/10.1016/j.biopha.2018.09.024
  12. Food Funct. 2018 Dec 07.
      Dihydroquercetin (DHQ), a dihydroxyflavone, possesses potent antioxidant properties and is proposed to be useful in the prevention and treatment of cardiovascular disease. In this study, we aimed to investigate whether DHQ has protective effects against MIRI and to elucidate the mechanism of attenuation of oxidative stress-and ERS-induced apoptosis via the PI3K/Akt pathway. Isolated rat hearts and H9c2 cardiomyocytes were treated with or without DHQ, and then subjected to I/R and H/R, respectively. Our results showed that DHQ pretreatment markedly alleviated cardiac dysfunction, scavenged free radicals, reduced lipid peroxidation, and increased the activity of antioxidant enzymes ex vivo and in vitro. The result of western blot analysis showed that DHQ inhibited the apoptotic pathway and the expression of pro-apoptotic proteins CHOP, Caspase-12, and p-JNK. In addition, DHQ delayed the onset of ERs by reducing GRP78, p-PERK, and p-eif2α expression levels and by increasing HO-1 expression and Nrf2 binding to antioxidant response elements. These cardioprotective effects of DHQ were partially counteracted by the PI3K/Akt inhibitor LY294002. The protective effects of DHQ on the inhibition of MIRI may be mediated by activating the PI3K/Akt pathway to reduce oxidative stress-and ERS-induced apoptosis.
    DOI:  https://doi.org/10.1039/c8fo01256c
  13. J Cell Biochem. 2018 Dec 12.
      Sevoflurane is the most commonly used general anesthetic in pediatric patients. But preclinical studies indicate that sevoflurane could have neurotoxicity in newborn and old animals, and this raises concern regarding its safety. In this study, we explored the potential mechanisms of sevoflurane-induced neurotoxicity in human SH-SY5Y neuronal cells. We showed that prolonged exposure to 2% sevoflurane caused a significant increase in the Bag family protein Bag2 in a time- and dose-dependent manner. We investigated the possible role of Bag2 upon exposure to sevoflurane by silencing Bag2 in neuronal cells. Knockdown of Bag2 caused increased overall reactive oxygen species (ROS) and generation of lipid peroxidation products 4-hydroxynonenal (4-HNE). Upon sevoflurane exposure, Bag2-silent cells have reduced glutathione (GSH) and glutathione peroxidase activity. Under the sevoflurane treatment, Bag2-deficient cells have reduced mitochondrial membrane potential (MMP) and adenosine triphosphate (ATP) production, while knockdown cells have less viability and higher lactic dehydrogenase (LDH) release as well as a higher percentage of apoptotic cells. The knockdown cells also had higher levels of mitochondrial cytochrome C release, a higher ratio of Bax/Bcl-2 and increased caspase cleavage by sevoflurane. Overall, our data support an important role of Bag2 in sevoflurane-induced neurotoxicity.
    Keywords:  Bag2; apoptosis; mitochondrial dysfunction; oxidative stress; sevoflurane
    DOI:  https://doi.org/10.1002/jcb.28029
  14. J Biosci. 2018 Dec;43(5): 857-865
      Oxidative stress and apoptosis is involved in hypoxia-reoxygenation (H/R) induced myocardial injury. Increased expression of uncoupling protein 2 (UCP2), a cationic carrier protein, has protective effect against H/R injury. The present study aimed to find candidate drugs for H/R induced cardiac damage by identifying compounds regulating UCP2 expression. Here, among six natural compounds, ursolic acid (UA) had the most significant induction effect on UCP2 expression in H9c2 cells under H/R conditions. Subsequently, we found that UA significantly attenuated cell apoptosis and Caspase 3 activity, but increased nitric oxide (NO) release under H/R conditions. Additionally, UA pretreatment also decreased reactive oxygen species (ROS) production and malondialdehyde (MDA) content, but increased superoxide dismutase (SOD) activity. H/R caused a notable increase in the phosphorylation of p38, which was weakened by UA pretreatment. Moreover, p38 inhibitor (SB203580) showed the similar effects on H/R cells as UA pretreatment, while UCP2 knockdown had the reverse biological effects. More importantly, the effects of UA or p38 inhibitor exposure were partially rescued by UCP2 knockdown. Collectively, our data suggested the functions of UA on UCP2 expression and on the protection of H/Rstimulated H9c2 cells may be attributed to p38 signaling pathway.
  15. J Exp Clin Cancer Res. 2018 Dec 12. 37(1): 309
      BACKGROUND: Prostate cancer is one of the most commonly diagnosed cancers in men worldwide. Currently available therapies for metastatic prostate cancer are only marginally effective. Therefore, new therapeutic agents are urgently needed to improve patient outcome. Isoalantolactone (IATL), an active sesquiterpene naturally present in many vegetables and medicinal plants, is known to induce cell death and apoptosis in various cancer cell lines. Nevertheless, antitumor mechanisms initiated by IATL in cancer cells have not been fully defined.METHODS: Cell apoptosis and cellular ROS levels were analyzed by flow cytometry. Western blot and qRT-PCR were used to analyze the protein and mRNA levels of indicated molecules, respectively. Nude mice xenograft model was used to test the effects of IATL on prostate cancer cell growth in vivo.
    RESULTS: In this study, we found that IATL dose-dependently inhibited cancer cell growth and induced apoptosis in PC-3 and DU145 cells. Mechanistically, our data found that IATL induced reactive oxygen species (ROS) production, resulting in the activation of endoplasmic reticulum stress pathway and eventually cell apoptosis in prostate cancer cells. IATL also decreased the protein expression levels of p-STAT3 and STAT3, and the effects of IATL were reversed by pretreatment with N-acetyl-L-cysteine (NAC). In vivo, we found that IATL inhibited the growth of prostate cancer xenografts without exhibiting toxicity. Treatment of mice bearing human prostate cancer xenografts with IATL was also associated with induction of ER stress and inhibtion of STAT3.
    CONCLUSION: In summary, our results unveil a previously unrecognized mechanism underlying the biological activity of IATL, and provide a novel anti-cancer candidate for the treatment of prostate cancer.
    Keywords:  ER stress; Isoalantolactone; Prostate cancer; Reactive oxygen species; STAT3
    DOI:  https://doi.org/10.1186/s13046-018-0987-9
  16. J Photochem Photobiol B. 2018 Dec 05. pii: S1011-1344(18)31013-3. [Epub ahead of print]190 146-153
      Zn-based phthalocyanine acts as drug or photosensitizer in photodynamic therapy (PDT) for the treatment of cancer cells. The activated zinc octacarboxyphthalocyanine (ZnPcOC) reacts with oxygen, to generate reactive oxygen species for the damage of melanoma cancer cells, Me45. This in vitro study aimed at investigating the cytotoxic effects of different concentrations of ZnPcOC activated with a diode laser (λ = 685 nm) on Me45, and normal human fibroblast cells, NHDF. To perform this study 104 cells/ml were seeded in 96-well plates and allowed to attach overnight, after which cells were treated with different concentrations of ZnPcOC (10, 20 and 30 μM). After 4 h, cells were irradiated with a constant light dose of 2.5; 4.5 and 7.5 J/cm2. Post-irradiated cells were incubated for 24 h before cell viability was measured using the MTT viability assay. Data indicated that high concentrations of ZnPcOC (30 μM) in its inactive state are not cytotoxic to the melanoma cancer cells and normal fibroblasts. Moreover, the results showed that photoactivated ZnPcOC (30 μM) was able to reduce the cell viability of melanoma and fibroblast to about 50%, respectively. At this photosensitizing concentration the efficacy the treatment light dose of 2.5; 4.5 and 7.5 J/cm2 was evaluated against Me45 cells. ZnPcOC at a concentration of 30 μM activated with a light dose of 2.5; 4.5 and 7.5 J/cm2 was the most efficient for the killing of melanoma cancer cells. Melanoma cancer cells after PDT with a photosensitizing concentration of 30 μM ZnPcOC and a treatment light dose of 2.5; 4.5 and 7.5 J/cm2 showed certain pro-apoptotic characteristics, such as direct inducer (early apoptosis) and long-term inducer, also (late apoptosis). This concludes that low concentrations of ZnPcOC, activated with the appropriate light dose, can be used to induce cell death in melanoma cells via ROS-induces apoptosis pathway, what was confirmed with cytometric ROS measurements. Our in vitro study showed that ZnPcOC mediated photodynamic therapy is an effective treatment option for melanoma Me45 cancer cells. 30 μM of ZnPcOC with the treatment light dose of 2.5 J/cm2 from a LED diode laser source, with a wavelength of 685 nm, was adequate to destroy melanoma cancer cells via ROS-induced apoptosis pathway, with low killing effects on healthy NHDF normal fibroblasts.
    Keywords:  Melanoma Me45 cancer cells; Photodynamic therapy (PDT); Photosensitizers; Pro-apoptotic activity; Reactive oxygen species (ROS); Zinc octacarboxyphthalocyanine (ZnPcOC), UV–Vis spectra
    DOI:  https://doi.org/10.1016/j.jphotobiol.2018.12.002
  17. J Cell Physiol. 2018 Dec 13.
      Osteosarcoma is a malignant primary bone tumor that responds poorly to both chemotherapy and radiation therapy. However, because of side effects and drug resistance in chemotherapy and the insufficiency of an effective adjuvant therapy for osteosarcoma, it is necessary to research novel treatments. This study was the first to investigate the anticancer effects of the flavonoid derivative artocarpin in osteosarcoma. Artocarpin induced cell apoptosis in three human osteosarcoma cell lines-U2OS, MG63, and HOS. Artocarpin was also associated with increased intracellular reactive oxygen species (ROS). Mitochondrial dysfunction was followed by the release of cytochrome c from mitochondria and accompanied by decreased antiapoptotic Bcl-2 and Bcl-xL and increased proapoptotic protein Bak and Bax. Artocarpin triggered endoplasmic reticulum (ER) stress, as indicated by changes in cytosol calcium levels and increased glucose-regulated protein 78 and 94 expressions, and also increased calpains expression and activity. Animal studies revealed a dramatic 40% reduction in tumor volume after 18 days of treatment. This study demonstrated a novel anticancer activity of artocarpin against human osteosarcoma cells and in murine tumor models. In summary, artocarpin significantly induced cell apoptosis through ROS, ER stress, mitochondria, and the caspase pathway, and may thus be a novel anticancer treatment for osteosarcoma.
    Keywords:  ER stress; ROS; apoptosis; artocarpin; osteosarcoma
    DOI:  https://doi.org/10.1002/jcp.27986
  18. Oncol Lett. 2018 Dec;16(6): 7131-7138
      Laryngeal squamous cell carcinoma (LSCC) is currently a serious public health problem in China; thus, it is urgent to identify effective treatment strategies for this disease. Previous studies demonstrated that reactive oxygen species (ROS) serve important roles in the apoptosis of LSCC cells. It has also been indicated that carboplatin (CBDCA), a second-generation platinum compound with broad antineoplastic properties, is able to induce oxidative stress to produce ROS, which in turn promotes apoptosis. Thus, the present study investigated if CBDCA is cytotoxic in LSCC cells due to the oxidative stress caused by ROS. Therefore, an MTT assay was performed to determine the cell viability of HN-3 LSCC cells following treatment with different doses of CBDCA. Subsequently, the expression levels of ROS and the rate of apoptosis/necrosis were evaluated in the cells. Following this, the HN-3 cells were co-treated with CBDCA and glutathione (GSH) or H2O2, followed by an MTT assay, a cell migration assay and western blot analysis. The results demonstrated that CBDCA reduced the viability of HN-3 cells in a time- and dose-dependent manner and promoted the production of ROS and apoptosis at certain doses. Additionally, the combination treatment of CBDCA and H2O2 enhanced the inhibitory effects of CBDCA on cell viability and migration ability, and promoted apoptosis in HN-3 cells; whereas the combined treatment of CBDCA and GSH exerted opposite effects. The results of the present study demonstrated that CBDCA promotes the apoptosis of HN-3 cells through accumulation of ROS, which may provide a novel treatment strategy for treating LSCC.
    Keywords:  apoptosis; carboplatin; laryngeal squamous cell carcinoma; oxidative stress; reactive oxygen species
    DOI:  https://doi.org/10.3892/ol.2018.9563
  19. J Agric Food Chem. 2018 Dec 09.
      Phloretin, a dihydrochalcone structural flavonoid compound, possesses antioxidant activity. In this study, we conducted studies to explore the function of phloretin on high palmitic acid-induced oxidative stress in human umbilical vein endothelial cells and investigated the potential mechanism using ribonucleic acid sequencing (RNA-Seq). Our findings reveal that phloretin significantly decreased the levels of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA), increased superoxide dismutase (SOD) and glutathione peroxidase-1 (Gpx-1) activity, and restored the loss of mitochondrial membrane potential (MMP). Next, whole transcriptome analysis was performed using RNA-Seq. The results indicated more than 3,000 differentially expressed genes (DEGs). Gene Ontology analysis revealed that the DEGs were categorized functionally mainly by the biological processes, cell metabolism, and cellular response to chemical stimulus. The Kyoto Encyclopedia of Genes and Genomes indicated that they were mainly enriched in cAMP, apoptosis, and cytoskeletal regulation signaling pathways. Furthermore, based on the results of RNA-Seq and western blotting, our study verified that phloretin upregulated the expression of p-Nrf2 and HO-1 by promoting the phosphorylation of AMPK at Thr172 through activation of liver kinase B1. In conclusion, phloretin attenuates PA-induced oxidative stress in HUVECs via the AMPK/Nrf2 anti-oxidative pathway.
    DOI:  https://doi.org/10.1021/acs.jafc.8b05025
  20. Iran J Basic Med Sci. 2018 Sep;21(9): 920-927
      Objectives: During type-1 diabetes treating by pancreatic islet transplantation, increasing oxidative stress and microbial contaminations are the main reasons of transplantation failure. In this study, we evaluated anti-apoptotic, antioxidant and antimicrobial potentials of phenolic compounds called ellagic acid (EA) and silybin on rat pancreatic islets.Materials and Methods: By doing MTT assay, effective concentrations of EA and silybin were determined as 1500 and 2100 μM, respectively. Then, ELISA methods, flow cytometry and MIC were done to investigate antioxidant, anti-apoptotic and antibacterial effects of those compounds, respectively.
    Results: Results of FITC Annexin-V and PI staining via flow cytometry, and also caspase-3 and -9 activities performed that EA has anti-apoptotic effects on pancreatic cells. Both compounds significantly diminished reactive oxygen species, and enhanced antioxidant power and insulin secretion. Furthermore, the minimum inhibitory concentration test indicated that these two have antibacterial effects on both Gram-positive and Gram-negative bacteria which usually contaminate the pancreatic islets.
    Conclusion: These findings support that use of EA and silybin can improve the function of islets which are used in transplantation, along with decreasing islets bacterial contamination.
    Keywords:  Antibacterial; Apoptosis; Ellagic acid; Islet transplantation Oxidative stress; Islets of Langerhans; Silybin
    DOI:  https://doi.org/10.22038/IJBMS.2018.27718.6753
  21. Med Sci Monit. 2018 Dec 11. 24 8970-8976
      BACKGROUND Resveratrol, a polyphenol found on the surface of red fruits, is able to suppress many kinds of malignancies. Nevertheless, its mechanism of action is not yet clear. Consequently, this study aimed to elucidate its influence and explore the etiology of PCCs (prostate cancer cells). MATERIAL AND METHODS The proliferation of prostate cancer cells was determined by CCK-8 assay. Cell apoptosis was determined by Hoechst staining FC assay. Cell migration was detected by scratch test. The levels of apoptosis-related protein were detected by Western blot analysis. RESULTS It was discovered that resveratrol suppresses cellular survival and migration and enhances cell death. In addition, it was revealed that resveratrol elevated ROS concentration and expression of biomarker of cell death Bax, while inhibiting Bcl2, an anti-apoptotic protein, and reinforcing expression of p53. Moreover, resveratrol remarkably increased the expressions of HIF-1α and p53 in PC cells. Resveratrol suppressed cell survival and promoted cell death, but its effects were reversed after HIF-1α knockdown, suggesting that the effects of resveratrol in PC are mediated via HIF-1α. CONCLUSIONS Our findings indicate that resveratrol induces apoptosis via HIF-1α/ROS/p53 signaling in prostate cancer cells and may be a useful therapeutic agent against prostate cancer.
    DOI:  https://doi.org/10.12659/MSM.913290
  22. Med Sci Monit Basic Res. 2018 Dec 10. 24 216-224
      BACKGROUND Curcumin has clear anti-tumor activity in various carcinomas. It regulates various signaling pathways like Wnt/β-catenin and JAK2/STAT3, which play vital roles in cell proliferation of several carcinomas, but to the best of our knowledge, there are currently no published reports on human glioma CHME cells. Therefore, the aim of this study was to explore the effect of curcumin on human glioma CHME cells. MATERIAL AND METHODS The CHME cell line was purchased from American Type Culture Collection (ATCC). The expressions of caspases 3, caspases 9, PARP, BAX, and BCL2 were detected by Western blot. Annexin V FITC, mitochondrial membrane potential, and reactive oxygen species were detected by flow cytometry. DAPI staining was detected by fluorescence microscopy. Cell viability was assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay. RESULTS We found that curcumin has cytotoxic activity in human glioma CHME cells, as shown by DAPI staining, annexin V/PI, and nuclear morphology. We found that cell growth decreased with increased concentration of curcumin, as well as sowing effects on expression of caspase-3, caspase-9, and cleavage of PARP, which suggests apoptotic cascade activity. The increase in reactive oxygen species and loss of mitochondrial membrane potential (Δψmt) in concentration-dependent manners suggests biochemical induction of apoptosis in CHME cells. CONCLUSIONS Curcumin has effective anticancer activity in human glioma CHME cells by inducing the apoptotic pathway.
    DOI:  https://doi.org/10.12659/MSMBR.912313
  23. Sci Total Environ. 2018 Nov 22. pii: S0048-9697(18)34662-X. [Epub ahead of print]656 778-788
      The brominated flame retardant, 2,2',4,4'-tetrabromodiphenyl ether (BDE-47), is well documented to exert potential negative impacts on different marine biota. However, the responsible mechanism remains unknown. The rainbow trout gonadal cell line RTG-2 was used as a model, and the mechanism and pathway underlying BDE-47 (6, 12.5 and 25 μM)-induced apoptosis and toxicity were examined in vitro. Apoptosis occurred in the RTG-2 cells exposed to BDE-47 in a clear concentration-dependent manner. The morphology of the mitochondrial alterations was observed using transmission electron microscopy. BDE-47 exposure decreased the cellular mitochondrial membrane potential, increased the cytochrome c released into the cytoplasm and elevated Fas protein expression. The mRNA expressions of Fas-associated death domain-containing protein (FADD), CHOP and GRP78 were also elevated, and similar increases were found in the activities of intracellular caspase-8, caspase-12, caspase-9 and caspase-3. These results indicated that the mitochondrial, endoplasmic reticulum and death-receptor pathways were involved in apoptosis in RTG-2 cells following BDE-47 exposure. ROS and Ca2+ were responsible for these changes because their overproduction was detected prior to apoptosis. However, the addition of the ROS scavenger N-acetyl-l-cysteine (NAC) and the intracellular calcium chelator (acetoxymethyl)-1,2-bis-(o-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM) did not significantly alleviate the apoptosis rate. The results of the present study show that BDE-47 exposure induced apoptosis in RTG-2 cells via ROS- and Ca2-mediated mitochondrial, endoplasmic reticulum and death-receptor apoptotic pathways.
    Keywords:  2,2′,4,4′-Tetrabromodiphenyl ether; Apoptotic pathway; Calcium; RTG-2 cells; Rainbow trout; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.scitotenv.2018.11.306
  24. Mol Med Rep. 2018 Nov 29.
      Although it is well reported that mitochondrial damage and endoplasmic reticulum (ER) stress (ERS) are involved in heavy metal‑induced cytotoxicity, the role of mitochondrial damage in hexavalent chromium [Cr(VI)]‑induced ERS and the correlation between the two have not been described and remain to be elucidated. The present study evaluated the ability of Cr(VI) to induce ERS in L‑02 hepatocytes, and subsequently examined the role of reactive oxygen species (ROS)‑mediated mitochondrial damage in Cr(VI)‑induced ERS. The findings demonstrated that Cr(VI) induced ERS, which was characterized by the upregulation of ERS‑associated genes and the substantial release of Ca2+ from the ER. The Cr(VI)‑induced mitochondrial production of ROS, by disturbing mitochondrial respiratory chain complexes I and II, may damage mitochondria directly by inducing mitochondrial permeability transition pore opening and mitochondrial membrane potential collapse. The results additionally demonstrated that Cr(VI) induced Ca2+ release from the ER through ROS/caveolin‑1/protein kinase B/inositol 1,4,5‑trisphosphate receptor signaling. The application of the ROS scavenger N‑acetyl‑cysteine confirmed the role of ROS in Cr(VI)‑mediated mitochondrial damage, ERS and apoptotic cell death. The data obtained demonstrated the role of mitochondrial damage in Cr(VI)‑induced ERS and provide novel insight into the elucidation of Cr(VI)‑induced cytotoxicity.
    DOI:  https://doi.org/10.3892/mmr.2018.9704
  25. J Cell Biochem. 2018 Dec 12.
      Oxidative stress and apoptosis in retinal pigment epithelium cells are involved in the pathogenesis of diabetic retinopathy (DR). Forkhead box class O 6 (FOXO6) is a member of the FOXO family that can regulate diabetes-induced oxidative stress. However, the role of FOXO6 in DR has not been clarified. The aim of the present study was to investigate the effects of FOXO6 on high glucose (HG)-induced oxidative stress and apoptosis in ARPE-19 cells. The results showed that FOXO6 was overexpressed in clinical vitreous samples from DR patients and in HG-induced ARPE-19 cells. Knockdown of FOXO6 by small interfeing RNA targeting FOXO6 (si-FOXO6) mitigated the HG-induced the production of reactive oxygen species and malondialdehyde, as well as the inhibition of superoxide dismutase activity. Knockdown of FOXO6 reduced the rate of cell apoptosis in HG-induced ARPE-19 cells. The increase in bax expression and decrease in bcl-2 expression caused by HG stimulation were reversed by si-FOXO6 transfection. Furthermore, knockdown of FOXO6 enhanced the activation of Akt/Nrf2 pathway in HG-stimulated ARPE-19 cells. Taken together, suppression of FOXO6 protects ARPE-19 cells from HG-induced oxidative stress and apoptosis, which is in part mediated by the activation of Akt/Nrf2 pathway.
    Keywords:  Akt/Nrf2 pathway; apoptosis; diabetic retinopathy; forkhead box class O 6; oxidative stress; retinal pigment epithelium cells
    DOI:  https://doi.org/10.1002/jcb.28252
  26. Andrologia. 2018 Dec 10. e13216
      This study evaluated the effects of the methanolic extract of Guibourtia tessmannii (GT) and selenium (Se) on cell viability, intracellular calcium concentration ([Ca2+ ]i ), apoptosis and oxidative stress through transient receptor potential vanilloid 1 (TRPV1) channel activity in CCL-97 (R2C) tumour Leydig cells. The cells were divided into nine groups and treated as follows: (a)-Control, (b)-Capsazepine (CPZ, 0.1 mM, a TRPV1 channel blocker), (c)-Capsaicin (CAP, 0.01 mM, a TRPV1 channel activator), (d)-GT (500 μg/ml), (e)-GT+CPZ, (f)-GT+CAP, (g)-Se (200 nM), (h)-Se+CPZ and (i)-Se+CAP. After treatments, cell viability, [Ca2+ ]i , apoptosis, caspase 3/9, reactive oxygen species (ROS) and mitochondrial membrane depolarisation (MMD) were evaluated. The [Ca2+ ]i , apoptosis, caspase 3/9, MMD and ROS levels were significantly (p < 0.001) increased in CAP group, but lowered in CPZ group. Interestingly, these parameters were significantly (p < 0.001) improved by GT and Se, compared to the CAP group. Moreover, the co-administration of GT+CAP or Se+CAP inhibited the cytotoxicity of CAP. Thus, the modulatory properties of GT and Se on Ca2+ influx, apoptosis and oxidative stress require the integrity of TRPV1 channel in CCL-97 Leydig cells. These results suggest that GT and Se might be used in the management of cytotoxicity in the testes, involving TRPV1 channel activity.
    Keywords:   Guibourtia tessmannii ; Leydig cells; Selenium; TRPV1 channel; oxidative stress
    DOI:  https://doi.org/10.1111/and.13216
  27. Toxicol Appl Pharmacol. 2018 Dec 06. pii: S0041-008X(18)30542-8. [Epub ahead of print]
      Radiation therapy toward malignancies is often ineffective owing to radioresistance of cancer cells. On the basis of anti-tumor properties of cordycepin, we examined the effects of cordycepin on sensitizing breast cancer cells toward radiotherapy. Cordycepin administration promoted G2/M arrest and apoptosis of MCF-7 and MDA-MB-231 cells resulting in restraining the proliferation of the cells in vitro and in vivo following irradiation. Mechanistic investigations showed that the breast cancer cells cultured with cordycepin harbored higher levels of intracellular reactive oxygen species (ROS) and incremental numbers of γ-H2AX foci after irradiation exposure. Importantly, cordycepin treatment down-regulated the expression levels of Nuclear factor erythroid 2-related factor (Nrf2) and a series of downstream genes, such as heme oxygenase-1 (HO-1), to enhance ROS in breast cancer cells exposed to irradiation. Together, our observations demonstrate that cordycepin treatment sensitizes breast carcinoma cells toward irradiation via Nrf2/HO-1/ROS axis. Thus, our findings provide novel insights into the function and the underlying mechanism of cordycepin in radiotherapy, and suggest that cordycepin might be employed as a radiosensitizer during radiotherapy toward breast cancer in a pre-clinical setting.
    Keywords:  Breast Cancer; Cordycepin; Nrf2; ROS; Radiosensitivity
    DOI:  https://doi.org/10.1016/j.taap.2018.12.006
  28. Biomed Pharmacother. 2018 Dec 04. pii: S0753-3322(17)37001-4. [Epub ahead of print]110 371-379
      BACKGROUND: Dendrobium candidum extract (DCE) has been reported to have anti-tumor property. However, the effect of DCE on liver cancer has not been well explored. This study was aimed to evaluate the inhibitory effect of DCE on liver cancer cells.METHODS: The effect of DCE on liver cancer cells was analyzed by detecting cell viability by MTT assay, detecting colony formation ability by colony formation assay, determining apoptotic cell rate, cell cycle distribution, active caspase-3 positive cells, Ki-67 positive cells, reactive oxygen species (ROS) level, and mitochondrial transmembrane potential (MMP) by flow cytometry analysis, and analyzing changes of expressions of cell cycle-, apoptosis-, and Wnt/β-catenin pathway-related proteins by Western blot.
    RESULTS: DCE inhibited viability and promoted apoptosis of liver cancer cell lines SMMC-7721 and BEL-7404. DCE decreased colony formation, induced cell cycle arrest, and led to cell cycle-associated proteins' abnormal expressions in SMMC-7721 and BEL-7404 cells. DCE effectively suppressed viability and proliferation of primary liver cancer cells and also induced aberrant expressions of cell cycle- and apoptosis-related proteins. After DCE treatment, ROS level was increased and MMP level was decreased. DCE inhibited β-catenin level in the nucleus and regulated downstream genes of β-catenin and further blocked Wnt/β-catenin pathway in SMMC-7721 and BEL-7404 cells as well as primary liver cancer cells.
    CONCLUSION: DCE suppressed liver cancer SMMC-7721 and BEL-7404 cells as well as primary liver cancer cells likely though activating mitochondria apoptosis pathway and inducing inhibition of Wnt/β-catenin pathway.
    Keywords:  Dendrobium candidum; Liver cancer; Mitochondria apoptosis; Wnt/β-catenin pathway
    DOI:  https://doi.org/10.1016/j.biopha.2018.11.149
  29. Food Chem Toxicol. 2018 Dec 07. pii: S0278-6915(18)30867-6. [Epub ahead of print]
      Since flavonoids are antioxidant compounds, they could beneficially affect neurodegenerative diseases where reactive oxygen species are involved. In this study, we firstly isolated and identified fourteen compounds from the flowers of Apios americana Medik. Then, we tested whether Apios americana Medik flowers water extract (AFWE) exerts a protective effect on H2O2 induced PC12 cells injure. As expected, pretreatment with AFWE inhibited cytotoxicity and DNA condensation in H2O2induced PC12 cells. Exposure of PC12 cells to H2O2 resulted in reactive oxygen species accumulation and mitochondrial dysfunction, while AFWE alleviated these damages. AFWE obviously reversed the alternations as H2O2 increased Caspase-3 and decreased the ratio of Bcl-2/Bax expressions. Furthermore, autophagy in PC12 cells was further activated by AFWE, which was beneficial to resisting adversity. These results manifest that AFWE prevents H2O2 induced damage via regulating autophagy.
    Keywords:  Apios americana medik flowers; Autophagy; Flavonoids; Oxidative stress
    DOI:  https://doi.org/10.1016/j.fct.2018.12.003
  30. Oxid Med Cell Longev. 2018 ;2018 3265918
      After spinal cord injury (SCI), some self-destructive mechanisms start leading to irreversible neurological deficits. It is known that oxidative stress and apoptosis play a major role in increasing damage after SCI. Metallothioneins I and II (MT) are endogenous peptides with known antioxidant, neuroprotective capacities. Taking advantage of those capacities, we administered exogenous MT to rats after SCI in order to evaluate the protective effects of MT on the production of reactive oxygen species (ROS) and lipid peroxidation (LP), as markers of oxidative stress. The activities of caspases-9 and -3 and the number of annexin V and TUNEL-positive cells in the spinal cord tissue were also measured as markers of apoptosis. Rats were subjected to either sham surgery or SCI and received vehicle or two doses of MT (10 μg per rat) at 2 and 8 h after surgical procedure. The results showed a significant increase in levels of MT protein by effect of SCI and SCI plus treatment at 12 h, while at 24 h an increase of MT was observed only in the injury plus treatment group (p < 0.05). ROS production was decreased by effect of MT in lesioned tissue; likewise, we observed diminished LP levels by MT effect both in the sham group and in the group with SCI. Also, the results showed an increase in the activity of caspase-9 due to SCI, without changes by effect of MT, as compared to the sham group. Caspase-3 activity was increased by SCI, and again, MT treatment reduced this effect only at 24 h after injury. Finally, the results of the number of cells positive to annexin V and TUNEL showed a reduction due to MT treatment both at 24 and 72 h after the injury. With the findings of this work, we conclude that exogenously administered MT has antioxidant and antiapoptotic effects after SCI.
    DOI:  https://doi.org/10.1155/2018/3265918
  31. Biotechnol Appl Biochem. 2018 Dec 13.
      The present study aimed to investigate the protective effects of salidroside (SAL) on 1-methyl-4-phenylpyridinium (MPP+ )-induced PC12 cell model for Parkinson's disease. PC12 cells were pretreated with SAL in different concentration and then exposed to MPP+ . To evaluate the effects of SAL on cytotoxicity, the survival rate was tested by MTT asssay and the apoptosis was tested via flow cytometry and western blot. ROS, GSH and MDA were detected to analysis the effects of SAL on oxidative stress. The mRNA and protein levels of inflammatory factors TNF-α and IL-1β were also determined by RT-qPCR and western blot. Pretreatment with SAL effectively relieved the MPP+ cytotoxic effects and decreased the release of ROS production and inflammatory cytokines. SAL also inhibited apoptosis, suppressed MDA activity and increased GSH levels in MPP+ -treated PC12 cells. Moreover, the expression levels of caspase-9, caspase-3 and Bax were significantly decreased in the SAL treatment groups compared with the MPP+ group, whereas Bcl-2 expression was significantly increased in the SAL treatment groups. In summary, the overall results suggested that SAL have neuroprotective effects on MPP+ -induced PC12 cell model by inhibiting inflammation, oxidative stress and cell apoptosis. SAL may be a potential active product to protect against Parkinson's disease. This article is protected by copyright. All rights reserved.
    Keywords:  Parkinson disease; apoptosis; inflammation; oxidative stress; salidroside
    DOI:  https://doi.org/10.1002/bab.1719
  32. Biochem Biophys Res Commun. 2018 Dec 10. pii: S0006-291X(18)32675-5. [Epub ahead of print]
      High glucose (HG) induces oxidative injury to cultured human umbilical vein endothelial cells (HUVECs). Recent studies have discovered 4-octyl itaconate (OI) as a novel and cell permeable Nrf2 (nuclear-factor-E2-related factor 2) activator. Its potential activity in HG-treated HUVECs was tested here. In HUVECs OI disrupted Keap1-Nrf2 association, causing Nrf2 protein accumulation and nuclear translocation, as well as transcription and expression of Nrf2-ARE-dependent genes, including HO1, NQO1 and GCLM. Significantly, pretreatment with OI potently inhibited HG (40 mM glucose)-induced death and apoptosis of HUVECs. Moreover, OI potently inhibited HG-induced reactive oxygen species (ROS) production, lipid peroxidation, superoxide accumulation and mitochondrial depolarization in HUVECs. Activation of Nrf2 is required for OI-induced cytoprotection in HUVECs. Nrf2 shRNA or knockout (by CRISPR/Cas9 method) reversed OI-mediated HUVEC protection against HG. Further studies showed that Keap1 silencing or Cys151S mutation mimicked and nullified OI-induced activity in HUVECs. Taken together, we conclude that OI activates Keap1-Nrf2 signaling to protect HUVECs from HG.
    Keywords:  4-octyl itaconate; HUVECs; High glucose; Nrf2 signaling; Oxidative injury
    DOI:  https://doi.org/10.1016/j.bbrc.2018.12.032
  33. Chemosphere. 2018 Dec 04. pii: S0045-6535(18)32295-1. [Epub ahead of print]219 227-235
      Arsenic (As) and copper (Cu) are ubiquitous environmental contaminants that are hazardous to the immune system. Our objective was to investigate the toxicity and potential mechanisms of thymus exposure to As and/or Cu. A chicken model was established by adding arsenic trioxide (As2O3; 30 mg/kg), copper sulfate (CuSO4; 300 mg/kg), and simultaneously both elements in the basal diet. After the chickens were fed for 12 weeks, a significant reduction in antioxidant enzyme levels or production of malondialdehyde (MDA) emphasized the occurrence of oxidative stress. Furthermore, an imbalance in mitochondrial dynamics along with its abnormal structure certified mitochondrial dysfunction. Additionally, elevated levels of pro-apoptotic and autophagy genes and decreased levels of antiapoptotic genes were found in treated groups. Karyopyknosis and chromatin peripheral condensation were accompanied by an increased apoptosis ratio, as well as accumulation of autophagosomes, thus indicating that apoptosis and autophagy are involved in immune cell death. All of the above thymus lesions and index abnormalities occurred in a time-dependent manner, and the Cu and As co-administered groups showed more deteriorating effects than the Cu and As groups alone. Moreover, in the As or Cu group, the thymus tissue suffered different susceptibilities in oxidative toxicity, which needs further study. Collectively, our results manifested that co-exposure to As and Cu increased the oxidative burden and exacerbated mitochondrial dysfunction on the thymus. Additionally, apoptosis and autophagy may act as partners in inducing cell death in a cooperative manner in chicken thymus after As and/or Cu exposure.
    Keywords:  Apoptosis; Arsenic; Autophagy; Copper; Mitochondrial dynamics; Oxidative stress
    DOI:  https://doi.org/10.1016/j.chemosphere.2018.11.188
  34. J Inorg Biochem. 2018 Nov 28. pii: S0162-0134(18)30471-9. [Epub ahead of print]191 174-182
      Six gallium(III) complexes (Ga1-Ga6) with 2‑quinolinecarboxaldehyde thiosemicarbazone analogues were synthesized and characterized. These gallium(III) complexes exhibited potent anticancer activity and exceeded that of the corresponding metal free ligands. Importantly, these gallium(III) complexes have a strong selectivity for tumor cells. Through the study of cellular mechanisms, we have found that the lipophilicity of ligands is closely linked to the antitumor activity of gallium(III) complexes. Additionally, we have chosen Ga6 with the best anti-tumor activity to study the mechanism of apoptosis. Caspase-3 and 9 activation and Annexin V-FITC/Propidium iodide (PI) dual-staining studies revealed that Ga6 promote apoptosis in A549 cells lines. Ga6 induces intracellular reactive oxygen species (ROS) and disrupts mitochondrial membrane potential.
    Keywords:  2‑Quinolinecarboxaldehyde thiosemicarbazone; Anticancer activity; Gallium(III) complex; Metallodrugs
    DOI:  https://doi.org/10.1016/j.jinorgbio.2018.11.017
  35. Iran J Basic Med Sci. 2018 Oct;21(10): 1013-1019
      Objectives: The purpose of this study was to detect the protective effects of adiponectin on coagulation dysfunction and its mechanism in sepsis of rats.Materials and Methods: The experimental samples were composed of sham group, model group that was underwent cecal ligation and puncture (CLP) and three adiponectin treatment groups that treated by adiponectin with different dose (72 μg/kg, 96 μg/kg and 120 μg/kg) after CLP. The prothrombin time (PT), activated partial thromboplastin time (APTT) was measured, respectively, the level of malondialdehyde (MDA), tissue factor (TF), activated coagulation factor VIIa and Xa, p-selectin were detected, the histology structure of vascular was observed, the expressions of Caspase 9, Caspase 3, Bax, Bcl-2 and vWF in vascular were measured.
    Results: The results demonstrated that adiponectin treatment lengthened PT and APTT, reduced the expression of MDA, TF, activated coagulation factor VIIa, Xa and p-selectin in plasma of septic rats. Additionally, adiponectin treatment alleviated endothelial cell apoptosis and oxidative stress, down-regulated the levels of Caspase 3, Caspase 9, Bax, Bcl-2 and vWF in vascular.
    Conclusion: These findings suggest that adiponectin treatment might be a promising therapeutic strategy for relieving septic endothelial cell injury and coagulation dysfunction via inhibiting endothelial cell apoptosis in septic rats.
    Keywords:  Adiponectin; Apoptosis; Endothelial cells; Oxidative stress; Rats; Sepsis; Thrombophilia
    DOI:  https://doi.org/10.22038/IJBMS.2018.29389.7117
  36. Med Sci Monit. 2018 Dec 12. 24 8993-9000
      BACKGROUND Exposure to ionizing radiation (IR) induces severe damage in multiple human tissues. The testes are extremely sensitive to IR, and testes irradiation can result in infertility and abnormality. A novel and safe radioprotector for testes injury from IR is needed. Polydatin (PD) has been proved to have anti-oxidant and anti-inflammatory effects, indicating its potential application in radiation protection. MATERIAL AND METHODS Male wild-type C57BL/6 mice (8 weeks old) were exposed to ionizing radiation. At different times after irradiation, testes were isolated and subjected to hematoxylin-eosin (HE) staining and TUNEL staining, as well as related quantification. ELISA assay was used to measure the level of inflammatory cytokines, and apoptosis proteins were detected by Western blot assay. Intracellular ROS was measured by DCFH-DA flow cytometry method. RESULTS In the present study, we demonstrated that polydatin effectively alleviated testes injury and retained sperm viability. PD pretreatment also inhibited cell apoptosis caused by irradiation. Radiation-induced decrease of FSH and testosterone was also inhibited by PD treatment. Finally, we showed that PD obviously reduced the ROS level, using DCFH-DA method. We also found that PD reduced the concentration of the oxidative products MDA and 8-OHdG. PD also inhibited apoptosis-related proteins such as Bax and caspase 3. CONCLUSIONS Our data proved that polydatin effectively alleviated testes injury after irradiation, mainly through reducing ROS and oxidative stress. Our findings suggest polydatin as a potential radioprotector for testes radiation damage.
    DOI:  https://doi.org/10.12659/MSM.913725
  37. Cell Physiol Biochem. 2018 ;51(5): 2341-2358
      BACKGROUND/AIMS: This study investigated the effect of consecutive superovulation on the ovaries and established a premature ovarian failure (POF) model in mice.METHODS: The mouse POF model was induced by 5-15 consecutive superovulation treatments with pregnant mare serum gonadotropin (PMSG), human chorionic gonadotropin (HCG) and prostaglandin F2α (PGF2α). Normal adult mice were compared with mice displaying natural ovarian aging. The following serum biochemical parameters were measured: including follicle-stimulating hormone (FSH), luteinizing hormone (LH), progesterone (P), estradiol (E2), inhibin B (INH B), malondialdehyde (MDA), total superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels. Follicles were counted using H&E staining. Levels of 8-hydroxyguanosine (8-OhdG), 4-hydroxynonenal (4-HNE), nitrotyrosine (NTY), anti-Mullerian hormone (AMH) and CDKN2A/ p16 (p16) were detected using immunohistochemical staining. Reactive oxygen species (ROS) levels were measured using dihydroethidium (DHE) staining. Cell apoptosis was detected using an in situ TUNEL fluorescence staining assay. Levels of proteins involved in ROS-related pathways and the p16 protein were detected using Western blotting. Sod1, Sod2 and Sod3 mRNA levels were detected using quantitative polymerase chain reaction (Q-PCR). Oocyte quality was evaluated using in vitro fertilization (IVF) and zygote culture.
    RESULTS: Consecutive superovulation groups presented lower P, E2, SOD, GSH-Px and INH B levels, significantly higher FSH, LH, MDA and ROS levels, and significantly fewer primordial follicles compared with the control group. Consecutive superovulation groups presented significantly increased levels of Sod2, 8-OhdG, 4-HNE, NTY, significantly increased levels of the SIRT1 and FOXO1 proteins, significantly increased levels of the senescence-associated protein p16, as well as decreased AMH, Sod1 and Sod3 levels and increased granulosa cell apoptosis compared with the control group.
    CONCLUSION: Consecutive superovulation significantly decreased ovarian function and oocyte quality and increased oxidative stress and apoptosis in the ovary via a mechanism involving the p16 and SIRT1/FOXO1 signaling pathways. These findings suggest that consecutive superovulation may be used to establish a mouse model of ovarian aging.
    Keywords:  Apoptosis; Ovary aging; Ovulation; Oxidative stress; Premature ovarian failure
    DOI:  https://doi.org/10.1159/000495895
  38. J Mol Neurosci. 2018 Dec 07.
      The objective of this study was to investigate the neuroprotective and antioxidant effects of ginsenoside compound K (CK) in a model of scopolamine hydrobromide-induced, memory-impaired mice. The role of CK in the regulation of amyloid β (Aβ) and its capacity to activate the Nrf2/Keap1 signaling pathway were also studied due to their translational relevance to Alzheimer's disease. The Morris water maze was used to assess spatial memory functions. Levels of superoxide dismutase, glutathione peroxidase, and malondialdehyde in brain tissues were tested. Cell morphology was detected by hematoxylin and eosin staining and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling assay. Immunohistochemistry and western blotting were used to determine expression levels of Nrf2/Keap1 signaling pathway-related factors and Aβ. Ginsenoside CK was found to enhance memory function, normalize neuronal morphology, decrease neuronal apoptosis, increase superoxide dismutase and glutathione peroxidase levels, reduce malondialdehyde levels, inhibit Aβ expression, and activate the Nrf2/Keap1 signaling pathway in scopolamine-exposed animals. Based on these results, we conclude that CK may improve memory function in scopolamine-injured mice by regulating Aβ aggregation and promoting the transduction of the Nrf2/Keap1 signaling pathway, thereby reducing oxidative damage to neurons and inhibiting neuronal apoptosis. This study suggests that CK may serve as a future preventative agent or treatment for Alzheimer's disease.
    Keywords:  Alzheimer’s disease; Amyloid β; Ginsenoside compound K; Scopolamine; Spatial memory
    DOI:  https://doi.org/10.1007/s12031-018-1210-3
  39. Mol Cell Probes. 2018 Dec 04. pii: S0890-8508(18)30285-8. [Epub ahead of print]
      Mitochondria play a central role in various critical cellular processes, including energy synthesis, energy supply and apoptosis. Panax notoginseng, a commonly used traditional Chinese medicine, has various pharmacological effects on the human body. Ginsenosides are representative bioactive components of P. notoginseng. Recently, more attention has focused on ginsenoside Rb1 as an antioxidative and anti-inflammatory agent that can protect the nervous system and the cardiovascular system. Numerous studies have shown that Rb1 exerts these effects by regulating mitochondrial energy metabolism, mitochondrial fission and fusion, apoptosis, oxidative stress and reactive oxygen species release, mitophagy and mitochondrial membrane potential. Thus, the mitochondria are pivotal targets of Rb1. This review summarized the available reports of the effects of ginsenoside Rb1 on the regulation of mitochondria and showed that it has a promising role in treating mitochondrial diseases.
    Keywords:  Apoptosis; Energy metabolism; Ginsenoside Rb1; MPTP; Mitochondria; Oxidative stress
    DOI:  https://doi.org/10.1016/j.mcp.2018.12.001
  40. Front Pharmacol. 2018 ;9 1347
      Patchouli alcohol (PA), a natural tricyclic sesquiterpene extracted from Pogostemon cablin (Blanco) Benth. (Labiatae), has been found to exhibit anti-Helicobacter pylori and anti-inflammatory properties. In this study, we investigated the protective effect of PA against H. pylori-induced gastritis in vitro and in vivo, and determined the underlying mechanism. In the in vivo experiment, a C57BL/6 mouse model of gastritis was established using H. pylori SS1, and treatments with standard triple therapy or 5, 10, and 20 mg/kg PA were performed for 2 weeks. Results indicated that PA effectively attenuated oxidative stress by decreasing contents of intracellular reactive oxygen species (ROS) and malonyldialdehyde (MDA), and increasing levels of non-protein sulfhydryl (NP-SH), catalase and glutathione (GSH)/glutathione disulphide (GSSG). Additionally, treatment with PA significantly attenuated the secretions of interleukin 1 beta (IL-1β), keratinocyte chemoattractant and interleukin 6 (IL-6). PA (20 mg/kg) significantly protected the gastric mucosa from H. pylori-induced damage. In the in vitro experiment, GES-1 cells were cocultured with H. pylori NCTC11637 at MOI = 100:1 and treated with different doses of PA (5, 10, and 20 μg/ml). Results indicated that PA not only significantly increased the cell viability and decreased cellular lactate dehydrogenase (LDH) leakage, but also markedly elevated the mitochondrial membrane potential and remarkably attenuated GES-1 cellular apoptosis, thereby protecting gastric epithelial cells against injuries caused by H. pylori. PA also inhibited the secretions of pro-inflammatory factors, such as monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor-α (TNF-α) and IL-6. Furthermore, after PA treatment, the combination of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) and cysteine-aspartic proteases 1 (CASPASE-1), the expression levels of NLRP3 inflammasome-related proteins, such as thioredoxin-interacting protein (TXNIP), pro-CASPASE-1, cle-CASPASE-1, and NLRP3 and genes (NLRP3 and CASPASE1) were significantly decreased as compared to the model group. In conclusion, treatment with PA for 2 weeks exhibited highly efficient protective effect against H. pylori-induced gastritis and related damages. The underlying mechanism might involve antioxidant activity, inhibition of pro-inflammatory factor and regulation of NLRP3 inflammasome function. PA exerted anti-H. pylori and anti-gastritis effects and thus had the potential to be a promising candidate for treatment of H. pylori-related diseases.
    Keywords:  Helicobacter pylori; gastric epithelial cell; gastritis; inflammasome; oxidative injury; patchouli alcohol
    DOI:  https://doi.org/10.3389/fphar.2018.01347
  41. Mol Cancer Res. 2018 Dec 14. pii: molcanres.0354.2018. [Epub ahead of print]
      Medullary thyroid carcinoma (MTC) originates from the C cells of the thyroid gland, which secrete calcitonin. Lymph node and distant metastases are frequently present at diagnosis. Activating mutations of RET, a driver oncogene in MTC that encodes a tyrosine kinase receptor, prevents apoptosis through inhibition of ATF4, a key transcriptional regulator of endoplasmic reticulum (ER) stress. We hypothesized that the combination of a tyrosine kinase inhibitor (TKI) and an ATF4 inducer promotes cell death by triggering catastrophic oxidative stress and apoptotic cell death. Here, we report that the ER-associated protein degradation (ERAD) inhibitor eeyarestatin sensitized MTC cells to the TKIs, sunitinib and vandetanib, thereby leading to synergistic upregulation of ATF4 expression, accumulation of reactive oxygen species, and subsequent cell death. Genome-wide analysis of ATF4 interaction sites by chromatin immunoprecipitation (ChIP) sequencing revealed that among ATF4 target genes was KLF9 (Kruppel like factor 9), which induces MTC apoptosis. ChIP assays revealed that ATF4 occupancy at the KLF9 promoter was increased in MTC cells treated with eeyarestatin or vandetanib alone and was further enhanced in cells treated with both drugs, leading to increased KLF9 transcription. Depletion of ATF4 by shRNA led to downregulation of KLF9 expression and prevented oxidative stress-induced cell death. Furthermore, we identified ATF4 target genes (LZTFL1, MKNK2, and SIAH1 with known tumor suppressor function) that were synergistically upregulated with the combination of TKI and ERAD inhibitor. Implications: These findings reveal a combination therapy that induces reactive oxygen species-dependent catastrophic cell death through induction of ATF4 and KLF9 transcriptional activity.
    DOI:  https://doi.org/10.1158/1541-7786.MCR-18-0354
  42. Oxid Med Cell Longev. 2018 ;2018 1526125
      The main purposes of this study were to screen the antioxidant activities of various fractions of Hemerocallis citrina Baroni and test their hepatoprotective effects in vitro. Antioxidant assays (2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), and reducing power experiments) and tert-butyl hydroperoxide- (t-BHP-) induced BRL-3A oxidative damage experiments were performed in vitro. The H. citrina ethyl acetate fraction (HCEA) was determined to have strong antioxidant activity because of its high flavonoid and polyphenol content. Ultraperformance liquid chromatography- (UPLC-) photodiode array (PDA)/mass spectrometry (MS) analysis showed that the main components of the HCEA were flavonoids and caffeic acid derivatives. A total of 17 compounds were identified. HCEA also effectively protected the liver against t-BHP-induced oxidative stress injury and significantly reduced reactive oxygen (ROS) accumulation. Moreover, HCEA significantly reduced levels of alanine aminotransferase (ALT), aspartate transaminase (AST), and lactate dehydrogenase (LDH). Further studies have shown that HCEA inhibits t-BHP-induced apoptosis by increasing B-cell lymphoma-2 (BCL-2) activity and decreasing caspase-3 and caspase-9 activity. Moreover, HCEA enhanced the activity of antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT), as well as the total antioxidant capacity (T-AOC), and increased the antioxidant level of glutathione (GSH) in BRL-3A cells. HCEA increased the antioxidant capacity of cells by increasing the gene expression of AMP-activated protein kinase (AMPK), extracellular signal-regulated kinase (ERK), P38, nuclear factor, erythroid 2 like 2 (Nrf2), SOD, glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modifier subunit (GCLM), and heme oxygenase 1 (HO-1), which are associated with antioxidant pathways to protect against oxidative stress. In conclusion, HCEA protected BRL-3A cells against t-BHP-induced oxidative stress damage via antioxidant and antiapoptosis pathways. Therefore, H. citrina Baroni may serve as a potential hepatoprotective drug.
    DOI:  https://doi.org/10.1155/2018/1526125
  43. Eur J Pharmacol. 2018 Dec 04. pii: S0014-2999(18)30699-X. [Epub ahead of print]844 65-72
      Fibroblast-like synoviocytes in rheumatoid arthritis (RA-FLSs) play a key role in cartilage destruction. We previously found that resveratrol (Res) could promote FLSs apoptosis in adjuvant arthritis rats, but the underlying mechanism was unclear. According to our latest study, Res can suppress the expression of mitochondrial superoxide dismutase (MnSOD) and RA-FLSs proliferation. It was associated with elevated mitochondrial reactive oxygen species levels. Therefore, we hypothesized that Res-mediated RA-FLSs apoptosis might occur via the MnSOD- mitochondrial reactive oxygen species pathway. RA-FLSs were infected with lentiviruses and screened with puromycin at a concentration of 8 µg/ml. We divided the RA-FLSs into four groups: a control group, a negative control (NC) group, a MnSOD overexpression group, and a MnSOD RNAi group. The four groups of RA-FLSs were tested using confocal laser scanning microscopy, CCK-8 assays, flow cytometry, and western blotting were conducted to determine the involvement of the MnSOD-mitochondrial reactive oxygen species pathway. Compared with the NC group, the MnSOD overexpression group treated with different concentrations of Res (0, 25, 50, 100, or 200 μM) and 5 μM H2O2 showed reduced levels of mitochondrial reactive oxygen species, increased B-cell-lymphoma-2 (Bcl-2), reduced Bcl-2 Associated X protein (Bax), and fewer apoptotic cells. The MnSOD RNAi group showed the opposite results. Thus, we concluded that Res could facilitate RA-FLSs apoptosis by regulating MnSOD expression and mitochondrial reactive oxygen species levels. Our findings show a novel mechanism for the beneficial effects of Res, especially in relation to the MnSOD-mitochondrial reactive oxygen species signaling pathway in RA.
    Keywords:  Apoptosis; Fibroblast-like synoviocytes; Lentivirus; MnSOD; Resveratrol
    DOI:  https://doi.org/10.1016/j.ejphar.2018.12.001
  44. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)36421-7. [Epub ahead of print]109 921-929
      Inhibition of histone deacetylase (HDAC) suppresses inflammation of pancreatic islets and apoptosis of β-cells. However, the underlying molecular mechanism is unclear. In the present study, we demonstrate that MGCD0103 (MGCD), an HDAC inhibitor, protects the pancreas from streptozotocin (STZ)-induced oxidative stress and cell death. Sprague-Dawley rats were intraperitoneally injected with STZ (40 mg/kg) to induce type I diabetes. MGCD (10 μg/day) was infused with osmotic mini-pump for 4 weeks. Pancreatic insulin and macrophage infiltration were analyzed by immunohistochemistry. Cellular level of reactive oxygen species (ROS) was evaluated with fluorescence-activated cell sorting. Tetramethylrhodamine ethyl ester was used to analyze mitochondrial membrane potential. Activation of caspase-3 was analyzed by western blotting. Chromatin immunoprecipitation was performed to investigate the binding affinity of specificity protein 1 (SP1) on the promoters of target genes. mRNA expression was analyzed by quantitative real-time polymerase chain reaction. As a result, we found that MGCD infusion ameliorated STZ-induced hyperglycemia, islet deformation, decreased insulin level, and macrophage infiltration. STZ injection promoted the production of ROS, which induced caspase activity and β-cell death. 4-Hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPOL), a mimetic of superoxide dismutase (SOD), reduced STZ-induced caspase activity and β-cell death. MGCD treatment increased SOD expression and histone acetylation level on promoters. Infusion of MGCD promoted acetylation of SP1 and its enrichment on SOD promoters. Thus, MGCD protects pancreatic β-cells from STZ-induced oxidative stress and cell death through the induction of antioxidant enzymes such as SODs.
    Keywords:  Histone deacetylase inhibitor; Oxidative stress; Pancreatic beta-cell death; Specificity protein 1; Type 1 diabetes
    DOI:  https://doi.org/10.1016/j.biopha.2018.10.163
  45. ACS Appl Mater Interfaces. 2018 Dec 12.
      In the field of nanomedicine, drug-loaded nanocarriers that integrate nanotechnology and chemotherapeutics are widely used to achieve synergistic therapeutic effects. Here we prepared mesoporous silica nanoparticles capped with cerium oxide nanoparticles (COP@MSN) where a pH triggered-responsive mechanism was used to control drug release and intracellular drug delivery. We blocked the mesopores of the carboxyl functionalized MSN with aminated COP. These pores could be open in acidic conditions to release the loaded drug, thus establishing a pH-responsive drug release system. We loaded doxorubicin (DOX) as a anticancer biomolecules into the pores of MSN and capped with COP. The COP@DOX-MSN system showed a typical drug release profile in an acidic medium, which however, was not observed in a neutral medium. In vitro studies using cancer cell line (HeLa) proved that the COP@DOX-MSN entered efficiently into HeLa cells and released DOX to the level sufficient for cytotoxicity. The cytotoxic effect of COP in cancer cells was facilitated by the pro-oxidant property of COPs, which considerably raised the reactive oxygen species (ROS) level, thereby leading to cellular apoptosis. The combination of DOX with COP (COP@DOX-MSN) showed even higher ROS level, demonstrating a cytotoxic synergism of drug and nanoparticle in terms of ROS generation. Collectively, the COP@DOX-MSN is considered useful for cancer treatment with the combined capacity of pH-controlled drug delivery, chemotherapeutics, and redox activity.
    DOI:  https://doi.org/10.1021/acsami.8b17958
  46. J Cell Physiol. 2018 Dec 06.
      Esophageal squamous cell carcinoma (ESCC) is one of the most common digestive tumors worldwide. The Mucin 1 (MUC1) heterodimeric protein has been confirmed that is overexpressed in ESCC and induced adverse outcomes. However, the detailed mechanism(s) remained challenging. So, we investigated the relationship between MUC1-C and metabolism in ESCC cells. In the results, TP53-induced glycolysis and apoptosis regulator (TIGAR) was overexpressed and correlative with MUC1-C positively in ESCC tissue. Targeting MUC1-C inhibits AKT-mTORC-S6K1 signaling and blocks TIGAR translation. We found that the inhibitory effect of GO-203 on TIGAR was mediated by inhibition of AKT-mTOR-S6K1 pathway. The findings also demonstrated that the suppressive effect of GO-203 on TIGAR is related to the decrease of glutathione level, the increase of reactive oxygen species and the loss of mitochondrial transmembrane membrane potential. In xenograft tissues, GO-203 inhibited the growth of ESCC cells and lead to the low expression of transmembrane C-terminal subunit (MUC1-C) and TIGAR. This evidence supports the contention that MUC1-C is significant for metabolism in ESCC and indicated that MUC1-C is a potential target for the treatment of ESCC.
    Keywords:  AKT; ESCC; GO-203; MUC1-C; TIGAR; metabolism
    DOI:  https://doi.org/10.1002/jcp.27863
  47. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)36092-X. [Epub ahead of print]109 726-733
      BACKGROUND: Subarachnoid hemorrhage (SAH) results in many brain dysfunctions and the neuroprotective function of puerarin after brain damage has been demonstrated in several studies. But whether puerarin can reduce brain nerve damage after SAH is not clear.In this study, we hypothesized that puerarin had the neuroprotective effect after SAH, and this protection could be mediated by bothBcl-2/Bax/Cleaved caspase-3 and SIRT3/SOD2 apoptotic signaling pathways.METHODS: First, we used neurological score, brain water content and so on to detect the neurological deficits after SAH. Then apoptosis neuron rate was detected by TUNEL staining. Western blot was carried out to explore the alteration of Blc-2, Bax, cleaved caspase-3 and Sirt3.Also, ROS acitivity and As-lysine level of SOD2 should be detected with assays.
    RESULTS: We demonstrate that puerarin attenuated the neurological deficits, effectively relieves cerebral edema, and reduce BBB disruption in SAH mice.And we revealed that a reduced rate of apoptosis neuron has been found out in puerarin treatment after SAH. In addition, obviously higher ratio of Blc-2/Bax and decreased expression of cleaved caspase-3 in puerarin-treated SAH micecomparing with vehicle-treated SAH animals had been found. Furthermore, puerarin effectively reversed these alterations in expression and inhibits ROSproduction induced by SAH. Also, puerarin can increase SOD activation after SAH and protect the expression of Sirt3 after SAH.
    CONCLUSIONS: In coclusion, puerarin can provide potential neuroprotection from the SAH damages, and can be act as a novel therapy for SAH.
    Keywords:  Puerarin; ROS; SIRT3; SOD2; Subarachnoid hemorrhage
    DOI:  https://doi.org/10.1016/j.biopha.2018.10.161
  48. Cancers (Basel). 2018 Dec 12. pii: E509. [Epub ahead of print]10(12):
      Targeted therapy is an effective, rational, and safe approach to solid and hematological tumors treatment. Unfortunately, a significant fraction of patients treated with tyrosine kinase inhibitors (TKI) relapses mainly because of gene amplification, mutations, or other bypass mechanisms. Recently a growing number of papers showed how, in some cases, resistance due to oncogene overexpression may be associated with drug addiction: cells able to proliferate in the presence of high TKI doses become also TKI dependent, undergoing cellular stress, and apoptosis/death upon drug withdrawal. Notably, if a sub-cellular population survives TKI discontinuation it is also partially re-sensitized to the same drug. Thus, it is possible that a subset of patients relapsing upon TKI treatment may benefit from a discontinuous therapeutic schedule. We focused on two different hematologic malignancies, chronic myeloid leukemia (CML) and anaplastic large cell lymphoma (ALCL), both successfully treatable with TKIs. The two models utilized (LAMA and SUP-M2) differed in having oncogene overexpression as the sole cause of drug resistance (CML), or additionally carrying kinase domain mutations (ALCL). In both cases drug withdrawal caused a sudden overload of oncogenic signal, enhanced mitochondria activity, induced the release of a high amount of reactive oxygen species (ROS), and caused genotoxic stress and massive cell death. In LAMA cells (CML) we could rescue the cells from death by partially blocking downstream oncogenic signaling or lowering ROS detrimental effect by adding reduced glutathione.
    Keywords:  cancer; inhibitors; oncogene; personalized medicine; targeted therapy
    DOI:  https://doi.org/10.3390/cancers10120509
  49. J Chem Neuroanat. 2018 Dec 07. pii: S0891-0618(18)30144-3. [Epub ahead of print]
      Ketamine is a dissociative anaesthetic agent whose recreational use amongst adolescents and young adults is reaching epidemic proportions in a number of countries. While animal studies have examined the long-term detrimental effects of early-life ketamine exposure, there is a paucity of information on the immediate effects of ketamine following subchronic administration in the adolescence period. Adolescent rats were assigned into four groups of 10 animals each, administered intraperitoneal (i.p) injections of vehicle or one of three doses of ketamine (7.5, 15 or 30 mg/kg daily) for 8 weeks, and then exposed to behavioural paradigms. Rats were then euthanised after an overnight fast, and blood taken was used for measurement of metabolic indices. The brains were dissected out and either homogenised for estimation of neurochemical parameters, or processed for histological and immunohistochemical studies. Results showed that subchronic administration of ketamine was associated with a lesser weight gain inspite of an increase in food intake across the treatment groups. There was a dose-dependent increase in open-field novelty-induced behaviours, a decline in spatial working-memory, and an anxiolytic effect in the elevated-plus maze. There was associated derangement of serum triglyceride, and increase in brain glutamate levels, acetylcholinesterase activity, plasma/brain oxidative stress parameters, caspase-3 activity and biochemical indices of hepatic and renal function. Ketamine administration was also associated with neurodegenerative changes in the cerebral cortex, hippocampus, cerebellum and the pons. In conclusion, subchronic administration of ketamine to adolescent rats was associated with dose-related memory loss, oxidative stress and possibly caspase-3 mediated neurodegenerative changes.
    Keywords:  Apoptosis; Excitotoxicity; Glial fibrillary acidic protein; Ketamine; Neurodegeneration; Oxidative stress
    DOI:  https://doi.org/10.1016/j.jchemneu.2018.12.002
  50. Cell Death Dis. 2018 Dec 11. 9(12): 1189
      Autophagy is a regulated, intracellular degradation process that delivers unnecessary or dysfunctional cargo to the lysosome. Autophagy has been viewed as an adaptive survival response to various stresses, whereas in other cases, it promotes cell death. Therefore, both deficient and excessive autophagy may lead to cell death. In this study, we specifically attempted to explore whether and how dysregulated autophagy contributes to caspase-dependent neuronal cell death induced by the neurotoxin 6-hydroxydopamine (6-OHDA). Ultrastructural and biochemical analyses indicated that MN9D neuronal cells and primary cultures of cortical neurons challenged with 6-OHDA displayed typical features of autophagy. Cotreatment with chloroquine and monitoring autophagic flux by a tandem mRFP-EGFP-tagged LC3 probe indicated that the autophagic phenomena were primarily caused by dysregulated autophagic flux. Consequently, cotreatment with an antioxidant but not with a pan-caspase inhibitor significantly blocked 6-OHDA-stimulated dysregulated autophagy. These results indicated that 6-OHDA-induced generation of reactive oxygen species (ROS) played a critical role in triggering neuronal death by causing dysregulated autophagy and subsequent caspase-dependent apoptosis. The results of the MTT reduction, caspase-3 activation, and TUNEL assays indicated that pharmacological inhibition of autophagy using 3-methyladenine or deletion of the autophagy-related gene Atg5 significantly inhibited 6-OHDA-induced cell death. Taken together, our results suggest that abnormal induction of autophagic flux promotes apoptotic neuronal cell death, and that the treatments limiting dysregulated autophagy may have a strong neuroprotective potential.
    DOI:  https://doi.org/10.1038/s41419-018-1229-y
  51. Natl J Maxillofac Surg. 2018 Jul-Dec;9(2):9(2): 160-166
      Introduction: Cancer of oral cavity is the uncontrolled expansion of damaged cell within the mouth cavity. 5-fluorouracil (5-FU) chemotherapy was focused to kill the cancer cell, but it would affect the surrounding normal cells during oral cancer treatment. This study included the evaluation of chemoprotective effects of curcumin (CU), as an herbal remedy, on 5-FU-induced-cytotoxicity toward oral cancer treatment, loaded within a nanocarrier system. CU was combined with 5-FU chemotherapy as a combinational drug-delivery system to evaluate synergistic effects.Materials and Methods: Nanoformulation of CU (nano-CU) and nanoformulation of 5-FU (nano-FU) were prepared by employing homogenization with high-energy sonication. The characterizations of prepared nanoformulations were evaluated on the basis of particle size, zeta potential, and polydispersity index (PDI) values. The chemopreventive effect of nano-CU on 5-FU induced-toxicity and synergistic efficacy were optimized through different in-vitro assays.
    Results: The average particle size of nano-CU and nano-FU were up to 200 nm, negatively-charged, and shown up to 4th-day control release of the drug within the acceptable concentration. IC50 value for growth inhibition was calculated as 47.89 and 26.19 μg/ml, respectively, for nano-CU and nano-FU. OCC was pretreated with nano-CU and shown the protective effect by reducing 5-FU induced-cytotoxicity by preventing normal cells through reduced viability. The DPPH-indicated fluorescence-tagged cells had quantified for antioxidant effect as it reduces intracellular reactive oxygen species level in OCC. Along with alteration in cell protein expression, Blc2, and Bax, shows enhanced apoptosis rate in OCC.
    Conclusion: Nano-CU provides chemoprotective nature towards 5-FU induced-toxicity, along with synergistic effects in oral cancer treatment.
    Keywords:  5-fluorouracil; cellular-toxicity; curcumin; nanocarrier; oral cancer
    DOI:  https://doi.org/10.4103/njms.NJMS_27_18
  52. Antioxidants (Basel). 2018 Dec 11. pii: E188. [Epub ahead of print]7(12):
      Skin cancer is among the most common cancer types accompanied by rapidly increasing incidence rates, thus making the development of more efficient therapeutic approaches a necessity. Recent studies have revealed the potential role of decosahexaenoic acid ester of phloridzin (PZDHA) in suppressing proliferation of liver, breast, and blood cancer cell lines. In the present study, we investigated the cytotoxic potential of PZDHA in an in vitro model of skin cancer consisting of melanoma (A375), epidermoid carcinoma (A431), and non-tumorigenic (HaCaT) cell lines. Decosahexaenoic acid ester of phloridzin led to increased cytotoxicity in all cell lines as revealed by cell viability assays. However, growth inhibition and induction of both apoptosis and necrosis was more evident in melanoma (A375) and epidermoid carcinoma (A431) cells, whereas non-tumorigenic keratinocytes (HaCaT) appeared to be more resistant as detected by flow cytometry. More specifically, PZDHA-induced cell cycle growth arrest at the G2/M phase in A375 and A431 cells in contrast to HaCaT cells, which were growth arrested at the G0/G1 phase. Elevated intracellular generation of reactive oxygen species ROS was detected in all cell lines. Overall, our findings support the potential of PZDHA as a novel therapeutic means against human skin cancer.
    Keywords:  apoptosis; cell cycle; docosahexaenoic acid; epidermoid carcinoma; flavonoids; melanoma; necrosis; oxidative stress; phloridzin; reactive oxygen species; skin cancer
    DOI:  https://doi.org/10.3390/antiox7120188
  53. Int J Mol Sci. 2018 Dec 13. pii: E4027. [Epub ahead of print]19(12):
      Morchella conica Pers. (M. conica) has been used both as a medical and edible mushroom and possesses antimicrobial properties and antioxidant activities. However, the antioxidant properties of polysaccharides purified from M. conica have not been studied. The aim of this study was to investigate the in vitro antioxidant properties of a polysaccharide NMCP-2 (neutral M. conica polysaccharides-2) purified from M. conica, as determined by radical scavenging assay and H₂O₂-induced oxidative stress in HEK 293T cells. Results showed that NMCP-2 with an average molecular weight of 48.3 kDa possessed a much stronger chelating ability on ferrous ions and a higher ability to scavenge radical scavenging 2,2-diphenyl-1-picrylhydrazyl (DPPH) than the other purified fraction of NMCP-1 from M. conica. Moreover, 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetra-zolium bromide (MTT) assay showed that NMCP-2 dose-dependently preserved cell viability of H₂O₂-induced cells. The NMCP-2 pretreated group reduced the generation of reactive oxygen species (ROS) content and increased the mitochondria membrane potential (MMP) levels. In addition, Hoechst 33342 staining revealed cells treated with NMCP-2 declined nuclear condensation. Ultrastructural observation revealed that NMCP-2 pretreatment alleviated the ruptured mitochondria when exposed to H₂O₂. Furthermore, western blot analysis showed that NMCP-2 prevented significant downregulation of the protein expression of Bax, cleaved caspases 3, and upregulated Bcl-2 levels. These results suggest the protective effects of NMCP-2 against H₂O₂-induced injury in HEK 293T cells. NMCP-2 could be used as a natural antioxidant of functional foods and natural drugs.
    Keywords:  Morchella conica; oxidative stress; polysaccharides
    DOI:  https://doi.org/10.3390/ijms19124027
  54. Comp Biochem Physiol C Toxicol Pharmacol. 2018 Dec 04. pii: S1532-0456(18)30169-8. [Epub ahead of print]217 106-113
      Tetrabromoethylcyclohexane (TBECH), as one emerging brominated flame retardants, is ubiquitous in the environment, including water and aquatic organisms. TBECH was found to exhibit endocrine-disrupting effects in different models, whereas a survey of comprehensive toxic effects of TBECH on zebrafish is limited. In the present study, zebrafish (Danio rerio) were waterborne exposed continuously to TBECH from embryonic stage (3 h post-fertilization (hpf)) to the time when the respective parameters were evaluated. Exposure to TBECH reduced hatchability of zebrafish embryos at 72 and 96 hpf, diminished heart rate of zebrafish larvae at 48 hpf, and increased malformation in zebrafish larvae at 96 hpf. In addition, exposure to TBECH diminished free swimming distance both in the light and under a photoperiod of 10 min light/10 min dark cycles in zebrafish larvae at 6 days post-fertilization (dpf). Moreover, exposure to TBECH elevated activities of superoxide dismutase (SOD) and catalase (CAT), malondialdehyde (MDA) content, whereas it reduced glutathione (GSH) content, in zebrafish larvae at 6 dpf. Accordingly, RT-qPCR analysis demonstrated that TBECH exposure increased the mRNA levels of sod1, sod2, cat, and gpx1 in zebrafish larvae at 6 dpf. With respect to the immune aspect, the mRNA levels of pro-inflammatory genes, including il-1b, il-6, il-8, and tnfa, in larval zebrafish at 6 dpf were increased by exposure to TBECH, while pretreatment with TBECH inhibited 24 h of exposure to LPS-stimulated elevation in the mRNA levels of the abovementioned four pro-inflammatory genes in zebrafish larvae at 6 dpf. Furthermore, TBECH treatment increased caspase-3 enzyme activities and regulated apoptosis-related genes in larval zebrafish at 6 dpf. Taken together, the data obtained in this study demonstrated that TBECH caused developmental and locomotor behavioral toxicity, immunotoxicity, oxidative stress and proapoptotic effects in early life zebrafish. The present study will help to understand the comprehensive toxicity of TBECH in zebrafish.
    Keywords:  Apoptosis; Developmental toxicity; Immunotoxicity; Oxidative stress; Tetrabromoethylcyclohexane; Zebrafish
    DOI:  https://doi.org/10.1016/j.cbpc.2018.12.004
  55. Front Mol Neurosci. 2018 ;11 432
      Light emitting diodes (LEDs) are widely used to provide illumination due to their low energy requirements and high brightness. However, the LED spectrum contains an intense blue light component which is phototoxic to the retina. Recently, it has been reported that blue light may directly impinge on mitochondrial function in retinal ganglion cells (RGCs). Mitochondria are high dynamic organelles that undergo frequent fission and fusion events. The aim of our study was to elucidate the role of mitochondrial dynamics in blue light-induced damage in retinal neuronal R28 cells. We found that exposure to blue light (450 nm, 1000 lx) for up to 12 h significantly up-regulated the expression of mitochondrial fission protein Drp1, while down-regulating the expression of mitochondrial fusion protein Mfn2 in cells. Mitochondrial fission was simultaneously stimulated by blue light irradiation. In addition, exposure to blue light increased the production of reactive oxygen species (ROS), disrupted mitochondrial membrane potential (MMP), and induced apoptosis in R28 cells. Notably, Drp1 inhibitor Mdivi-1 and Drp1 RNAi not only attenuated blue light-induced mitochondrial fission, but also alleviated blue light-induced ROS production, MMP disruption and apoptosis in cells. Compared with Mdivi-1 and Drp1 RNAi, the antioxidant N-acetyl-L-cysteine (NAC) only slightly inhibited mitochondrial fission, while significantly alleviating apoptosis after blue light exposure. Moreover, we examined markers for mitophagy, which is responsible for the clearance of dysfunctional mitochondria. It was found that blue light stimulated the conversion of LC3B-I to LC3B-II as well as the expression of PINK1 in R28 cells. Mdivi-1 or Drp1 RNAi efficiently inhibited the blue light-induced expression of PINK1 and co-localization of LC3 with mitochondria. Thus, our data suggest that mitochondrial fission is required for blue light-induced mitochondrial dysfunction and apoptosis in RGCs.
    Keywords:  apoptosis; blue light; mitochondrial fission; mitophagy; retinal neuronal cells
    DOI:  https://doi.org/10.3389/fnmol.2018.00432
  56. Oncol Rep. 2018 Dec 07.
      Gadolinium (Gd) compounds serve as magnetic resonance imaging contrast agents and exert certain anticancer activities. Yet, the molecular signaling underlying the antitumor effect of Gd chloride (GdCl3) on glioma remains unclear. In the present study, we aimed to ascertain the apoptotic mechanisms of GdCl3 on rat glioma C6 cells. Our results demonstrated that GdCl3 significantly reduced cell viability and shrunk cell morphology of C6 cells in a concentration‑dependent manner. GdCl3 led to apoptotic C6 cell death as detected by TUNEL staining. An increase in cleaved caspase‑3, cleaved caspase‑8 and cleaved caspase‑9 occurred in GdCl3‑treated C6 cells as detected by immunoblotting analysis. The activities of caspase‑3, caspase‑8 and caspase‑9 were increased, and the specific inhibitors of caspase‑3/‑8/‑9 individually reversed cell viability, which caused apoptotic death in C6 cells prior to GdCl3 exposure. GdCl3 also caused an elevation in the cytoplasmic Ca2+ level and reactive oxygen species (ROS) production, as well as the loss of mitochondrial membrane potential (ΔΨm) as shown by flow cytometric analysis in C6 cells. The results from the immunoblotting analysis demonstrated that there were upregulated protein levels of cytochrome c and Bax but a downregulated protein level of Bcl‑2 in C6 cells after GdCl3 treatment. Additionally, GdCl3 decreased the protein levels of phosphorylated‑extracellular signal‑regulated kinases, phosphorylated‑c‑Jun N‑terminal kinase and phosphorylated‑p38 mitogen‑activated protein kinases in C6 cells. In conclusion, ROS production and MAPKs signaling pathways contribute to GdCl3‑induced caspase cascade‑mediated apoptosis in C6 cells. Our findings provide a better understanding of the molecular mechanisms underlying the role of GdCl3 in rat glioma C6 cells.
    DOI:  https://doi.org/10.3892/or.2018.6913
  57. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)34696-1. [Epub ahead of print]109 2054-2061
      Hepatocellular carcinoma (HCC) is one of the most common cancers and has a high mortality rate in less developed countries, especially in China. Galangin (GA), one of the most important and naturally active flavonoids, extracted primarily from the root of Alpinia officinarum Hance, has been demonstrated to be effective in the treatment of HCC. It is a substance with defensive actions and a broad range of biological properties, including inhibitory effects on bacteria, fungi, viruses, the control of hypertension and diabetes, and chemoprevention of several cancers. Experiments have shown that GA prevents HCC through multiple anti-cancer mechanisms, anti-genotoxic activity against environmental and dietary carcinogens; anti-proliferative effects through reversal of the Warburg effect in HCC; arrest of the cell cycle in the G0/G1 phase; induction of apoptosis via stimulation of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and the mitochondrial-dependent apoptosis pathway; induction of autophagy; and inhibition of angiogenesis, metastasis, and multidrug resistance (MDR). In addition, synergistic effects with other chemotherapy drugs have been demonstrated. Therefore, this review is focused on the anti-HCC mechanisms of GA.
    Keywords:  Apoptosis; Autophagy; Chemopreventive effects; Galangin; Hepatocellular carcinoma
    DOI:  https://doi.org/10.1016/j.biopha.2018.09.154
  58. Nucleic Acids Res. 2018 Dec 12.
      The tissue specificity of mitochondrial tRNA mutations remains largely elusive. In this study, we demonstrated the deleterious effects of tRNAThr 15927G>A mutation that contributed to pathogenesis of coronary artery disease. The m.15927G>A mutation abolished the highly conserved base-pairing (28C-42G) of anticodon stem of tRNAThr. Using molecular dynamics simulations, we showed that the m.15927G>A mutation caused unstable tRNAThr structure, supported by decreased melting temperature and slower electrophoretic mobility of mutated tRNA. Using cybrids constructed by transferring mitochondria from a Chinese family carrying the m.15927G>A mutation and a control into mitochondrial DNA (mtDNA)-less human umbilical vein endothelial cells, we demonstrated that the m.15927G>A mutation caused significantly decreased efficiency in aminoacylation and steady-state levels of tRNAThr. The aberrant tRNAThr metabolism yielded variable decreases in mtDNA-encoded polypeptides, respiratory deficiency, diminished membrane potential and increased the production of reactive oxygen species. The m.15927G>A mutation promoted the apoptosis, evidenced by elevated release of cytochrome c into cytosol and increased levels of apoptosis-activated proteins: caspases 3, 7, 9 and PARP. Moreover, the lower wound healing cells and perturbed tube formation were observed in mutant cybrids, indicating altered angiogenesis. Our findings provide new insights into the pathophysiology of coronary artery disease, which is manifested by tRNAThr mutation-induced alterations.
    DOI:  https://doi.org/10.1093/nar/gky1241
  59. J Am Chem Soc. 2018 Dec 13.
      Nanoformulations that can respond to the specific tumor microenvironment (TME), such as a weakly acidic pH, low oxygen, and high glutathione (GSH), show promise for killing cancer cells with minimal invasiveness and high specificity. In this study, we demonstrate self-assembled copper-amino acid mercaptide nanoparticles (Cu-Cys NPs) for in situ glutathione-activated and H2O2-reinforced chemodynamic therapy for drug-resistant breast cancer. After endocytosis into tumor cells, the Cu-Cys NPs could first react with local GSH, induce GSH depletion and reduce Cu2+ to Cu+. Subsequently, the generated Cu+ would react with local H2O2 to generate toxic hydroxyl radicals (•OH) via a Fenton-like reaction, which has a fast reaction rate in the weakly acidic TME, that are responsible for tumor-cell apoptosis. Due to the high GSH and H2O2 concentration in tumor cells, which sequentially trigger the redox reactions, Cu-Cys NPs exhibited relatively high cytotoxicity to cancer cells, whereas normal cells were left alive. The in vivo results also proved that Cu-Cys NPs efficiently inhibited drug-resistant breast cancer without causing obvious systemic toxicity. As a novel copper mercaptide nanoformulation responsive to the TME, these Cu-Cys NPs may have great potential in chemodynamic cancer therapy.
    DOI:  https://doi.org/10.1021/jacs.8b08714
  60. Curr Oral Health Rep. 2018 ;5(4): 211-220
      Purpose of Review: Oral health is maintained in a dynamic equilibrium between the host immunity and the oral microbiome. Oral polymorphonuclear neutrophils (oPMNs) are important innate immune cells in the oral cavity.Recent Findings: The oPMNs play a co-controlling part in the maintenance of oral equilibrium. In human saliva, the oPMNs integrity is preserved, and their function remains unaffected. In general, oPMNs are in a higher state of baseline activation compared to peripheral PMNs. However, in periodontitis, the oPMNs' activation state can result in excessive release of damaging molecules in the extracellular environment.
    Summary: The presence of oPMNs may unwittingly negatively impact the integrity of the oral tissues. While most of the oPMN functions occur intracellularly, release of their potent active mediators into the extracellular environment may jeopardize oral homeostasis and its integrity. The dual nature of oPMNs, both beneficial and detrimental, remains a challenging and understudied topic.
    Keywords:  Apoptosis; Degranulation; Neutrophils; Oral health; Reactive oxygen species
    DOI:  https://doi.org/10.1007/s40496-018-0199-6
  61. Oncol Rep. 2018 Oct 29.
      Ursodeoxycholic acid (UDCA) is a type of hydrophilic bile acid extracted from animal bile with a wide range of biological functions. The present results demonstrated that UDCA could effectively inhibit the proliferation of two human melanoma cell line (M14 and A375) with time‑ and concentration‑dependence. Following exposure to various concentrations of UDCA, M14 cells exhibited typical morphological changes and weaker ability of colony forming. Flow cytometry analysis demonstrated that UDCA could induce a decrease of mitochondrial membrane potential and an increase in reactive oxygen species (ROS) levels in M14 cells. The cell cycle was arrested in the G2/M phase, which was confirmed by the decrease of cyclin‑dependent kinase 1 and cyclinB1 at the protein level. However, when M14 cells were treated with UDCA and Z‑VAD‑FMK (caspase inhibitor) synchronously, the apoptosis rate of the cells was reduced significantly. In addition, it was demonstrated that UDCA induced apoptosis of human melanoma M14 cells through the ROS‑triggered mitochondrial‑associated pathway, which was indicated by the increased expression of cleaved‑caspase‑3, cleaved‑caspase‑9, apoptotic protease activating factor‑1, cleaved‑poly (ADP‑ribose) polymerase 1 and the elevation of B cell lymphoma‑2 (Bcl‑2) associated X protein/Bcl‑2 ratio associated with apoptosis. Therefore, UDCA may be a potential drug for the treatment of human melanoma.
    DOI:  https://doi.org/10.3892/or.2018.6828
  62. Methods Mol Biol. 2019 ;1894 1-29
      As a consequence of their increase in annual production and widespread distribution in the environment, nanoparticles potentially pose a significant public health risk. The sought-after catalytic activity granted by their physiochemical properties doubles as a hazard to physiological processes following exposure through inhalation, oral, transdermal, subcutaneous, and intravenous uptake. Upon uptake into the body, their size, morphology, surface charge, coating, and chemical composition augment the response of biological systems to the materials and enhance their toxicity. Identification of each property is necessary to predict the harm imposed by foreign nanomaterials in the body. Assay methods ranging from endotoxin and lactate dehydrogenase (LDH) signaling to apoptosis and oxidative stress detection supply valuable techniques for exposing biomarkers of nanoparticle-induced cellular damage. Spectroscopic investigation of epithelial barrier permeation and distribution within living cells reveals the proclivity of nanoparticles to penetrate the body's natural defensive boundaries and deposit themselves in cytotoxic locations. Combination of the various characterization methodologies and assays is required for every new nanoparticulate system despite preexisting data for similar systems due to the lack of deterministic trends among investigated nanoparticles. The propensity of nanomaterials to denature proteins and oxidize substrates in their local environment generates significant concern for the applicability of several traditional in vitro assays, and the modification of susceptible approaches into novel methods suitable for the evaluation of nanoparticles comprises the focus of future work centered on nanoparticle toxicity analysis.
    Keywords:  Biocompatibility; In vitro assays; Mechanisms of toxicity; Nanomaterials; Nanoparticle characterization; Nanoparticle toxicity; Nanoparticle-cell interaction; Reactive oxygen species
    DOI:  https://doi.org/10.1007/978-1-4939-8916-4_1
  63. Front Oncol. 2018 ;8 548
      Since resistance to radiotherapy remains refractory for the clinical management of nasopharyngeal cancer (NPC), further understanding the mechanisms of radioresistance is necessary in order to develop more effective NPC treatment and improve prognosis. In this study, an integrated quantitative proteomic approach involving tandem mass tag labeling and liquid chromatograph-mass spectrometer was used to identify proteins potentially responsible for the radioresistance of NPC. The differential radiosensitivity in NPC model cells was examined through clonogenic survival assay, CCK-8 viability assay, and BrdU incorporation analysis. Apoptosis of NPC cells after exposure to irradiation was detected using caspase-3 colorimetric assay. Intracellular reactive oxygen species (ROS) was detected by a dichlorofluorescin diacetate fluorescent probe. In total, 5,946 protein groups were identified, among which 5,185 proteins were quantified. KEGG pathway analysis and protein-protein interaction enrichment analysis revealed robust activation of multiple biological processes/pathways in radioresistant CNE2-IR cells. Knockdown of MAPK15, one up-regulated protein kinase in CNE2-IR cells, significantly impaired clonogenic survival, decreased cell viability and increased cell apoptosis following exposure to irradiation, while over-expression of MAPK15 promoted cell survival, induced radioresistance and reduced apoptosis in NPC cell lines CNE1, CNE2, and HONE1. MAPK15 might regulate radioresistance through attenuating ROS accumulation and promoting DNA damage repair after exposure to irradiation in NPC cells. Quantitative proteomic analysis revealed enormous metabolic processes/signaling networks were potentially involved in the radioresistance of NPC cells. MAPK15 might be a novel potential regulator of radioresistance in NPC cells, and targeting MAPK15 might be useful in sensitizing NPC cells to radiotherapy.
    Keywords:  MAPK15; nasopharyngeal carcinoma; quantitative proteomics; radioresistance; radiosensitivity
    DOI:  https://doi.org/10.3389/fonc.2018.00548
  64. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)34668-7. [Epub ahead of print]109 886-891
      Piceatannol (3,3',4,5'-trans-trihydroxystilbene) is a natural polyphenols compound that occurs hydroxylated analogue of resveratrol showing widely biological activities. Previous studies have demonstrated its functions on anti-cancer, neuroprotection and cardioprotection. However, few studies have clarified the benefits of piceatannol on cardiomyocytes except its anti-oxidative effect based on the original property of polyphenols. Here we apply H9c2 cardiomyocytes to study the cardioprotective mechanisms of piceatannol in vitro. We firstly verify its anti-peroxidation effect by using H2O2-induced in vitro model. Then, flow cytometry results show piceatannol reduce cellular apoptosis by enhancing Bcl-2 expressions in immunoblot analysis. Meantime, piceatannol decreases H2O2-induced excessive ROS and calcium overloading, and prevents mitochondrial depolarization. Most importantly, piceatannol pretreatment can regulate PI3K-Akt-eNOS signaling pathway to alleviate peroxidative injury. Immunoblot analysis of PI3K, Akt, p-Akt and eNOS shows H2O2 significantly reduces expressions of these proteins. Pretreatment of piceatannol evidently increases their expressions and decreases iNOS expression, implying piceatannol can upregulate PI3K-Akt-eNOS signaling to protect cardiomyocytes from peroxidative injury.
    Keywords:  Bcl-2; PI3K-Akt-eNOS; Peroxidative injury; Piceatannol
    DOI:  https://doi.org/10.1016/j.biopha.2018.10.120
  65. Neuropsychiatr Dis Treat. 2018 ;14 3111-3119
      Background: Intracerebral hemorrhage (ICH) is a devastating neurological injury associated with significant mortality. Necroptosis is a newly identified type of programmed necrosis initiated by the activation of tumor necrosis factor alpha. Evidences had demonstrated the importance of necroptosis in neuronal cell death. Necrostatin-1 is a specific inhibitor of necroptosis. The present study was carried out to explore whether RIP1/RIP3 pathways participate in hemin induced cell death in HT-22 hippocampal neuronal cells and investigate the potential neuroprotection of necrostatin-1 in hemin induced cell death in HT-22.Methods: First, different concentrations of hemin (0, 25, 50, 100 μmol/L) were added to HT-22 cells. Propidium iodide (PI) positive cells and cell viability were measured at 24 hours after hemin treatment. Then, necrostatin-1, pan-caspase inhibitor Benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone (z-VAD-fmk) and reactive oxygen species (ROS) scavenger butylated hydroxyanisole (BHA) were applied to hemin-treated HT-22 cells. PI positive cells and cell viability were measured at 24 hours after hemin treatment. MitoSox Red was used to indicate ROS level. Last, the effect of RIP3 in hemin induced HT-22 cell death was explored through RIP3 knockdown using siRNA. PI positive cells, cell viability and ROS lever were measured at 24 h after hemin treatment.
    Results: Hemin could induce a dose dependent cell death in HT22 neural cells. RIP1 specific inhibitor necrostatin-1 significantly inhibited cell death induced by hemin in HT-22 cells, greatly reducing PI positive cells, dramatically improving cell viability and decreasing ROS accumulation. BHA could significantly inhibit PI positive cells induced by hemin in HT-22 cells. Furthermore, silencing of RIP3 using siRNA attenuated hemin induced cell death in HT-22 cells, greatly reducing PI positive cells, dramatically improving cell viability and decreasing ROS accumulation.
    Conclusion: These data revealed that RIP1/RIP3 might mediate hemin induced cell death in HT-22 cells, and necrostatin-1 played a neuroprotection role in hemin induced cell death in HT-22. RIP1 and RIP3 might represent novel therapeutic targets for ICH.
    Keywords:  HT22; RIP1; RIP3; intracerebral hemorrhage; necroptosis; necrostatin-1
    DOI:  https://doi.org/10.2147/NDT.S181074
  66. Front Pharmacol. 2018 ;9 1334
      Oxidative stress has been highlighted as therapeutic targets for acetaminophen (APAP)-induced hepatotoxicity. Isoorientin (Iso), a well-known flavonoid-like compound, has been shown to have antioxidant potential. However, the effect of Iso on APAP-induced liver injury has not yet been elucidated. The present study investigated the hepatoprotective effect of Iso and its underlying mechanism. C57BL/6J mice were used to evaluate the hepatoprotective effect of Iso in vivo and HepG2 cells were utilized to further decipher the mechanisms of Iso -induced Nrf2 activation. We found that Iso treatment significantly reduced APAP-induced hepatotoxicity by reducing the lethality, histopathological liver changes, and alanine transaminase (ALT) and aspartate aminotransferase (AST) levels in serum. These effects were accompanied by decreased malondialdehyde (MDA) formation and myeloperoxidase level (MPO), and by decreased superoxide dismutase (SOD) and glutathione (GSH) depletion. Moreover, Iso induced Nrf2 activation and translocation as well as upstream AMPK/Akt/GSK3β activation. Furthermore, Iso effectively alleviated mitochondrial dysfunction by reducing c-jun N-terminal kinase phosphorylation and translocation, Bax mitochondrial translocation, and apoptosis-inducing factor and cytochrome c release. Further mechanistic investigations revealed that the activation of Nrf2 by Iso via the AMPK/Akt/GSK3β pathway contributed to the hepatoprotective activity of Iso in vitro. In addition, the Iso-mediated inhibition of APAP-induced the lethality, histopathological changes and mitochondrial dysfunction observed in WT mice was nearly absent in Nrf2-/- mice. In summary, Iso ameliorated APAP-induced hepatotoxicity by activating Nrf2 via the AMPK/Akt/GSK3β pathway.
    Keywords:  Isoorientin Iso; Nrf2; acetaminophen APAP; hepatotoxicity; oxidative stress
    DOI:  https://doi.org/10.3389/fphar.2018.01334
  67. Theriogenology. 2018 Dec 03. pii: S0093-691X(18)30555-7. [Epub ahead of print]126 55-62
      The aim of this study was to investigate effects of nitric oxide (NO) on steroidogenesis and apoptosis in goat luteinized granulosa cells (LGCs). We cultured goat LGCs from healthy follicles in culture medium supplemented with the NO donor sodium nitroprusside (SNP) or the NO synthase inhibitor Nω-Nitro-l-arginine methyl ester hydrochloride (l-NAME), then examined steroid synthesis, oxidative stress and apoptosis in vitro. The results showed that SNP treatment significantly increased the cGMP concentration in the LGCs (P < 0.05), whereas the l-NAME treatment significantly decreased cGMP concentration (P < 0.05). Then Inhibition of NO production significantly inhibited the expression of CYP19A1, a key gene that is involved in sex steroid hormones synthesis and is responsible for the decrease of E2. Inhibition of NO production resulted in an increased percentage of apoptosis, which was accompanied by upregulating expression levels of apoptosis-related markers BAX, CASP3 and CASP9. These data indicate that NO is required for goat LGCs steroidogenesis and cell survival. Furthermore, Inhibition of NO production decreased the expression of mitochondrial biogenesis related genes and proteins (PPARGC1A, NRF-1 and TFAM) and the mtDNA copy number. Simultaneously, inhibition of NO production suppressed the transcription and translation of SOD, GPX1, and CAT, and decreased the glutathione level and increased the 8-OHdG level. However, SNP treatment increased the expression of genes involved in mitochondrial function and biogenesis, and elevated the anti-oxidant stress system and steroid synthesis. Together, our results indicate that NO may up-regulate the expression of PPARGC1A and its downstream factors through the cGMP pathway, thereby decreasing granulosa cell apoptosis, and may participate in the regulation of granulocyte steroid production through the mitochondrial-dependent pathway.
    Keywords:  Apoptosis; Goat luteinized granulosa cells; Mitochondria; Nitric oxide; Steroidogenesis
    DOI:  https://doi.org/10.1016/j.theriogenology.2018.12.007
  68. Gene. 2018 Dec 06. pii: S0378-1119(18)31242-3. [Epub ahead of print]
      Moringa oleifera (Moringaceae) is a plant known for having high antioxidant potency, anticancer, hepatoprotective, cardioprotective etc. and many more activities. Besides these, Moringaceae has the potential for attenuating the male sexual dysfunction. Reactive oxygen species/ROS were increased in cryptorchidism and therefore cause infertility by damaging sperm DNA and germ cell apoptosis. There was an increase in heat shock proteins (HSP) in cells, which is affected by heat shock. In the present study, the antioxidant effects of two different doses of M. oleifera Lam Extract (MOLE) on experimentally induced cryptorchid testes of rats was investigated. Forty two male rats (16 days old) were divided into four groups: a normal control group, a cryptorchidism-induced control group and two cryptorchidism-induced groups treated orally with either 400 or 800 mg/kg MOLE for 2 weeks. Our study showed that there were ruptures from interstitial spaces, separation of the germ cells from basal membrane, falling of the germ cells into the lumen, perivascular fibrosis, oedema, increased level of HSP70, apoptosis, malondialdehyde (MDA) and decrease in the level of superoxide dismutase (SOD) after the cryptorchidism. We found that pathological damages, oxidative stress, expression of the HSP70 and germ cell apoptosis were decreased in treated groups with MOLE. In brief, we can say that aqueous extract of M. oleifera reduces the oxidative stress in a unilateral cryptorchidism induced rats, and it might attenuate histopathological damages, HSP expression and germ cell apoptosis.
    Keywords:  Apoptosis; Cryptorchidism; HSP70; Male infertility; Moringa oleifera; Oxidative stress
    DOI:  https://doi.org/10.1016/j.gene.2018.11.091
  69. Biochem Pharmacol. 2018 Dec 08. pii: S0006-2952(18)30506-9. [Epub ahead of print]
      Pharmaceuticals and their by-products are increasingly a matter of concern, because of their unknown impacts on human health and ecosystems. The lack of information on these transformation products, which toxicity may exceed that of their parent molecules, makes their detection and toxicological evaluation impossible. Recently we characterized the Pyridinium of furosemide (PoF), a new transformation product of furosemide, the most widely used diuretic and an emerging pollutant. Here, we reveal PoF toxicity in SH-SY5Y cells leading to alpha-synuclein accumulation, reactive oxygen species generation, and apoptosis. We also showed that its mechanism of action is mediated through specific inhibition of striatal respiratory chain complex I, both in vitro by direct exposure of striatum mitochondria to PoF, and in vivo, in striatal mitochondria isolated from mice exposed to PoF for 7 days in drinking water and sacrificed 30 days later. Moreover, in mice, PoF induced neurodegenerative diseases hallmarks like phospho-Serine129 alpha-synuclein, tyrosine hydroxylase decrease in striatum, Tau accumulation in hippocampus. Finally, we uncovered PoF as a new metabolite of furosemide present in urine of patients treated with this drug by LC/MS. As a physiopathologically relevant neurodegeneration inducer, this new metabolite warrants further studies in the framework of public health and environment protection.
    Keywords:  Alpha-synuclein; Metabolite; Mitochondrial complex I; Neurodegenerative diseases; Pharmaceutical pollutant; ROS
    DOI:  https://doi.org/10.1016/j.bcp.2018.12.007
  70. Biochim Biophys Acta Mol Cell Res. 2018 Dec 05. pii: S0167-4889(18)30172-1. [Epub ahead of print]
      The prevailing models of stress induced Programmed Cell Death (PCD) posit that excess extracellular chemicals interact with or enter cells and disrupts cellular homeostasis. This activates signalling cascades involving the mitochondria, an increase in the steady state levels of Reactive Oxygen Species (ROS) as well as the activation of Bax and caspases. Further, the increased ROS also causes cellular damage that triggers or enhances PCD responses. The models have been modified in a number of ways, for example to include the existence of caspase and Bax independent forms of PCD. More recently, the ubiquity of ROS has also been challenged in part based on the failure of anti-oxidants to protect from diseases with increased intensity of oxidative stress. Here we focus on a number of other, often overlooked, observations regarding stress mediated responses that may further increase our mechanistic understanding of PCD. These include the concept of the "milieu intérieur" which suggests that cells actively protect themselves (adaptive homeostasis) in part by limiting entry to most extracellular chemicals. Of similar importance, stress also increases the levels of other stress inducible second messengers including ceramide, iron and calcium. This review focuses on the concept that stress is an agonist that conveys information that is transduced into the cell to activate the appropriate genetically encoded cell death and survival responses.
    DOI:  https://doi.org/10.1016/j.bbamcr.2018.12.001
  71. Cell Stress. 2017 Oct;1(1): 11-36
      This review presents current knowledge related to VDAC1 as a multi-functional mitochondrial protein acting on both sides of the coin, regulating cell life and death, and highlighting these functions in relation to disease. It is now recognized that VDAC1 does not only play a crucial role in regulating the metabolic and energetic functions of mitochondria. The location of VDAC1 at the outer mitochondrial membrane (OMM) allows the control of metabolic cross-talk between mitochondria and the rest of the cell and also enables its interaction with proteins involved in metabolic and survival pathways. Along with regulating cellular energy production and metabolism, VDAC1 is also involved in the process of mitochondria-mediated apoptosis by mediating the release of apoptotic proteins and interacting with anti-apoptotic proteins. VDAC1 functions in the release of apoptotic proteins located in the mitochondrion inter-membranal space via oligomerization to form a large channel that allows passage of cytochrome c and AIF and their release to the cytosol, subsequently apoptotic cell death. VDAC1 also regulates apoptosis via interactions with apoptosis regulatory proteins, such as hexokinase (HK), Bcl2 and Bcl-xL, some of which are also highly expressed in many cancers. This review also provide insight into VDAC1 function in Ca2+ homeostasis, oxidative stress, and presents VDAC1 as a hub protein interacting with over 100 proteins. Such interactions enable VDAC1 to mediate and regulate the integration of mitochondrial functions with cellular activities. VDAC1 can thus be considered as standing at the crossroads between mitochondrial metabolite transport and apoptosis and hence represents an emerging cancer drug target.
    Keywords:  Apoptosis; Cancer; Metabolism; Mitochondria; VDAC1
    DOI:  https://doi.org/10.15698/cst2017.10.104
  72. Biomed Pharmacother. 2019 Jan;pii: S0753-3322(18)34525-6. [Epub ahead of print]109 815-822
      Arsenic was an established carcinogen and toxicant, occurring in drinking water and food. Arsenic was increasingly being blamed as a risk factor for diabetes mellitus. Recent studies have found that arsenic could induce the generation of reactive oxygen species (ROS) and mitochondria were the major targets of ROS. Damage mitochondria could be removed by mitophagy and mitophagy played a defensive role against cellular apoptosis. To investigate whether the arsenic could induce the injury in mitochondria, we treated Wistar rat offsprings and INS-1 cells with As2O3 and sodium arsenite, respectively. Our results showed that arsenic induced the generation of ROS in both rat offsprings' pancreas and INS-1 cells. The generation of ROS induced by arsenic could inhibit the expression of PPARγ. PPARγ is a major impact on mitochondrial function. The inhibition of PPARγ induced the reduction of PINK1 signaling and the upregulation of Bax. PINK1 signaling was one of the classical pathways of mitophagy. The inhibition of mitophagy induced the activation of apoptosis both in rat offsprings' pancreas and INS-1 cells. After treated with Rosiglitazone (RGS, PPARγ receptor agonist), PPARγ was rescued, the expression of PINK1 significantly increasing and the apoptosis was restrained. We used Taurine (Tau) as the protective agent both in rat offsprings' pancreas and INS-1 cells, after treated with Tau, the production of ROS was decreased significantly and the downgrade of PPARγ was rescued.
    Keywords:  Apoptosis; Arsenic; Mitophagy; PPARγ
    DOI:  https://doi.org/10.1016/j.biopha.2018.10.134
  73. FEBS Open Bio. 2018 Dec;8(12): 1936-1942
      Diabetic retinopathy is the most important ocular complication associated with diabetes. Corneal defects due to diabetes mellitus (DM) may cause severe vision impairments. This study aimed to identify the effect of transforming growth factor-β (TGF-β) on biological events, such as apoptosis and inflammation, in the diabetic cornea. High-glucose treatment induced reactive oxygen species (ROS) production and several biological events, including apoptosis and inflammatory cytokine secretion, in human corneal epithelial cells. However, administration of TGF-β significantly decreased ROS production, Annexin V-positive cells, and levels of inflammatory cytokines. Sprague Dawley rats were injected with streptozotocin (STZ) as a model of DM. Inflammatory cytokine secretion, apoptosis, and inflammation were all increased by STZ treatment. However, apoptosis and inflammation were markedly reduced following TGF-β treatment. In conclusion, TGF-β can ameliorate the enhancement of apoptosis and inflammation in diabetic cornea in in vivo and in vitro.
    Keywords:  apoptosis; cornea; glucose; inflammation; transforming growth factor‐β
    DOI:  https://doi.org/10.1002/2211-5463.12529
  74. Mol Cancer Res. 2018 Dec 14. pii: molcanres.0931.2018. [Epub ahead of print]
      Acquired drug resistance remains a challenge in chemotherapy. Here we show enzymatic, in-situ assembling of cholesterol derivatives to act as polypharmaceuticals for selectively inducing death of cancer cells via multiple pathways and without inducing acquired drug resistance. A conjugate of tyrosine and cholesterol (TC), formed by enzyme catalyzed dephosphorylation of phosphorylate TC (pTC), self-assembles selectively on or in cancer cells. Acting as polypharmaceuticals, the assemblies of TC augment lipid rafts, aggregate extrinsic cell death receptors (e.g., DR5, CD95, or TRAILR), modulate the expression of oncoproteins (e.g., Src and Akt), disrupt the dynamics of cytoskeletons (e.g., actin filaments or microtubules), induce ER stress, and increase the production of reactive oxygen species (ROS), thus resulting in cell death and preventing acquired drug resistance. Moreover, the assemblies inhibit the growth of platinum-resistant ovarian cancer tumor in a murine model. This work illustrates the use of instructed-assembly (iA) in cellular environment to form polypharmaceuticals in-situ that not only interact with multiple proteins, but also modulate membrane dynamics for developing novel anticancer therapeutics. Implications: As a multifaceted strategy for controlling cancer cell death, instructed-assembly (iA) minimized acquired resistance of cancer cells, which is a new strategy to amplify the genetic difference between cancer and normal cells and provides a promises for overcoming drug resistance in cancer therapy.
    DOI:  https://doi.org/10.1158/1541-7786.MCR-18-0931
  75. J Stroke Cerebrovasc Dis. 2019 Jan;pii: S1052-3057(18)30523-8. [Epub ahead of print]28(1): 175-184
      BACKGROUND: Stevioside, isolated from the herb Stevia rebaudiana, has been widely used as a food sweetener all over the world. Isosteviol Sodium (STV-Na), an injectable formulation of isosteviol sodium salt, has been proved to possess much greater solubility and bioavailability and exhibit protective effects against cerebral ischemia injury in vivo by inhibiting neuron apoptosis. However, the underlying mechanisms of the neuroprotective effects STV-Na are still not completely known. In the present study, we investigated the effects of STV-Na on neuronal cell death caused by hypoxia in vitro and its underlying mechanisms.METHODS: We used cobalt chloride (CoCl2) to expose mouse neuroblastoma N2a cells to hypoxic conditions in vitro.
    RESULTS: Our results showed that pretreatment with STV-Na (20 μM) significantly attenuated the decrease of cell viability, lactate dehydrogenase release and cell apoptosis under conditions of CoCl2-induced hypoxia. Meanwhile, STV-Na pretreatment significantly attenuated the upregulation of intracellular Ca2+ concentration and reactive oxygen species production, and inhibited mitochondrial depolarization in N2a cells under conditions of CoCl2-induced hypoxia. Furthermore, STV-Na pretreatment significantly downregulated expressions of nitric oxide synthase, interleukin-1β, tumor necrosis factor-α, interleukin-6, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) signalings in N2a cells under conditions of CoCl2-induced hypoxia.
    CONCLUSIONS: Taken together, STV-Na protects neural cells against hypoxia-induced apoptosis through inhibiting MAPK and NF-κB pathways.
    Keywords:  Cerebral ischemia; STV-Na; cell apoptosis; cobalt chloride; hypoxia; mitogen-activated protein kinase
    DOI:  https://doi.org/10.1016/j.jstrokecerebrovasdis.2018.09.020
  76. Iran J Basic Med Sci. 2018 Sep;21(9): 896-904
      Objectives: Heat stress (HS) is a catastrophic stressor that dampens immunity. The current study investigates the effect of dietary administration with camel whey protein (CWP) on apoptotic pathway caused by HS.Materials and Methods: Forty-five male mice were divided into three groups: a control group; HS group; and HS mice that were orally supplemented with CWP (CWP-HS group).
    Results: We found that reactive oxygen species (ROS), pro-inflammatory cytokines (IL-6), and C reactive protein (CRP) were elevated in the HS group along with a significant increase of caspase-9 and -3 and decrease of total antioxidant capacity (TAC). HS mice revealed impaired phosphorylation of Bcl-2 and Survivin, as well as increased expression of Bax, Bim and cytochrome C. Additionally, we observed an aberrant distribution of HSP-70 expressing lymphocytes in the spleen and thymus of HS mice. Moreover, histopathological examination showed alterations on the architectures of immune organs. In comparison with CWP-HS group, we found that CWP restored the levels of ROS, IL-6, TAC and CRP induced by HS. Furthermore, CWP restored the expression of Bcl-2/Bax, improved the histopathological changes in immune organs and HSP-70 distribution in the spleen and thymus.
    Conclusion: Our findings revealed the possible ameliorative role of CWP supplementation against damages induced by exposure to HS.
    Keywords:  Antioxidants; Apoptosis; Camel whey protein; Free radicals; Heat stress
    DOI:  https://doi.org/10.22038/IJBMS.2018.27584.6729
  77. BMB Rep. 2018 Dec 14. pii: 4455. [Epub ahead of print]
      Aging is accompanied by a time-dependent progressive deterioration of multiple factors of the cellular system. The past several decades have witnessed major leaps in our understanding of the biological mechanism of aging using dietary, genetic, pharmacological, and physical interventions. Metabolic processes, including nutrient sensing pathways and mitochondrial function, have emerged as prominent regulators of aging. Mitochondria have been considered to play a key role largely due to their production of reactive oxygen species (ROS), resulting in DNA damage that accumulates over time and ultimately causes cellular failure. This theory, known as the mitochondrial free radical theory of aging (MFRTA), was favored by the aging field, but increasing inconsistent evidence has led to criticism and rejection of this idea. However, MFRTA should not be hastily rejected in its entirety because we now understand that ROS is not simply an undesired toxic metabolic byproduct, but also an important signaling molecule that is vital to cellular fitness. Notably, mitochondrial function, a term traditionally referred to bioenergetics and apoptosis, has since expanded considerably. It encompasses numerous other key biological processes, including the following: (i) complex metabolic processes, (ii) intracellular and endocrine signaling/communication, and (iii) immunity/inflammation. Here, we will discuss shortcomings of previous concepts regarding mitochondria in aging and their emerging roles based on recent advances. We will also discuss how the mitochondrial genome integrates with major theories on the evolution of aging.
  78. Int J Mol Sci. 2018 Dec 09. pii: E3964. [Epub ahead of print]19(12):
      The p38 MAPK pathway is known to influence the anti-tumor effects of several chemotherapeutics, including that of organometallic drugs. Previous studies have demonstrated the important role of p38 both as a regulator and a sensor of cellular reactive oxygen species (ROS) levels. Investigating the anti-cancer properties of novel 1,8-naphthalimide derivatives containing Rh(I) and Ru(II) N-heterocyclic carbene (NHC) ligands, we observed a profound induction of ROS by the complexes, which is most likely generated from mitochondria (mtROS). Further analyses revealed a rapid and consistent activation of p38 signaling by the naphthalimide-NHC conjugates, with the Ru(II) analogue-termed MC6-showing the strongest effect. In view of this, genetic as well as pharmacological inhibition of p38α, attenuated the anti-proliferative and pro-apoptotic effects of MC6 in HCT116 colon cancer cells, highlighting the involvement of this signaling molecule in the compound's toxicity. Furthermore, the influence of MC6 on p38 signaling appeared to be dependent on ROS levels as treatment with general- and mitochondria-targeted anti-oxidants abrogated p38 activation in response to MC6 as well as the molecule's cytotoxic- and apoptogenic response in HCT116 cells. Altogether, our results provide new insight into the molecular mechanisms of naphthalimide-metal NHC analogues via the ROS-induced activation of p38 MAPK, which may have therapeutic interest for the treatment of various cancer types.
    Keywords:  N-heterocyclic carbene; ROS; apoptosis; cancer; mitochondria; naphthalimide-metal complex conjugates; p38 MAPK
    DOI:  https://doi.org/10.3390/ijms19123964