bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒03‒10
83 papers selected by
Gavin McStay
Staffordshire University


  1. J Chin Med Assoc. 2019 Feb;82(2): 92-98
      BACKGROUND: Halofuginone, which is the main active ingredient of Dichroa fabrifuga, was used to inhibit the synthesis of type I collagen and played increasingly important roles in tumor therapy. This study aims to investigate the protective effects of halofuginone on human umbilical vein endothelial cells (HUVECs) from H2O2-induced apoptosis and oxidative stress.METHODS: Propidium iodide and Annexin-V double staining assay was used to measure the apoptosis. Cell viability assay, the measurements of reactive oxygen species (ROS) parameters malondialdehyde and superoxide dismutase, western-blot assays, and quantitative PCR were used to elucidate the effects and mechanisms of halofuginone in protecting H2O2-induced injury.
    RESULTS: The results showed that halofuginone counteracted H2O2-induced cell viability decline and PCNA downregulation. Furthermore, halofuginone decreased ROS levels and protected HUVECs from H2O2-induced apoptosis. In detail, it showed that H2O2 induced a transient activation of Mitogen-activated protein kinases members ERK1/2 and p38, whereas induced a sustained activation of c-Jun N-terminal kinase (JNK), which play dominant roles in triggering apoptosis. Inhibition of JNK activation also inhibited H2O2-mediated apoptosis. Finally, it was shown that halofuginone upregulated VEGF expressions, which functioned by inhibiting sustained JNK activation, thus protecting HUVECs.
    CONCLUSION: Halofuginone has powerful effects in protecting HUVECs from H2O2-induced apoptosis, via upregulating VEGF and inhibiting overactivated JNK phosphorylation. Halofuginone might be a promising preventive drug for cardiovascular diseases.
    DOI:  https://doi.org/10.1097/JCMA.0000000000000008
  2. J Toxicol Sci. 2019 ;44(3): 155-165
      Silver nanoparticles (AgNPs) are increasingly utilized in a number of applications. This study was designed to investigate AgNPs induced cytotoxicity, oxidative stress and apoptosis in rat tracheal epithelial cells (RTE). The RTE cells were treated with 0, 100 μg/L and 10,000 μg/L of the AgNPs with diameters of 10 nm and 100 nm for 12 hr. The cell inhibition level, apoptosis ratio, reactive oxygen species (ROS), malondialdehyde (MDA) and metallothionein (MT) content were determined. The mRNA expression of cytoc, caspase 3, and caspase 9 was measured by quantitative real-time polymerase chain reaction (qRT-PCR). In addition, we also analyzed the cytoc, caspase 3, pro-caspase 3, caspase 9, and pro-caspase 9 protein expression by western blotting. Electric cell-substrate impedance sensing (ECIS) analysis showed that the growth and proliferation of RTE cells were significantly inhibited in a dose-dependent manner under AgNPs exposure. The cell dynamic changes induced by 10 nm AgNPs were more severe than that of the 100 nm AgNPs exposure group. The intracellular MT, ROS, and MDA content increased when the exposure concentration increased and size reduced, whereas Ca2+-ATPase activity and Na+/K+-ATPase activity changed inversely. The relative expression of protein of cytoc, caspase 3, and caspase 9 were upregulated significantly, which indicated that AgNPs induced apoptosis of RTE cells through the caspase-dependent mitochondrial pathway. Our results demonstrate that AgNPs caused obvious cytotoxicity, oxidative stress, and apoptosis in RTE cells, which promoted the releasing of cytochrome C and pro-apoptotic proteins into the cytoplasm to activate the caspase cascade and finally led to apoptosis.
    Keywords:  Apoptosis; Cytotoxicity; Oxidative stress; Rat tracheal epithelial cells; Silver nanoparticles
    DOI:  https://doi.org/10.2131/jts.44.155
  3. Arch Toxicol. 2019 Mar 08.
      Autophagy and apoptosis are the major types of cell death in pesticide-induced neurotoxicity, and autophagy is known to play a role in cell protection by inhibiting apoptosis. In this study, we characterized the relationship between autophagy and apoptosis in diquat (DQ)-induced cell death and explored a novel pharmacotherapeutic approach involving autophagy regulation to prevent DQ neurotoxicity. DQ was cytotoxic to PC12 cells in a concentration-dependent manner, as shown by decreased cell viability and decreased dopamine (DA) levels. DQ-induced apoptosis was found in PC12 cells, as demonstrated by activation of caspase-3 and -9 and by nuclear condensation. By monitoring expression of microtubule-associated protein 1A/1B light chain 3B (LC3-II) and p62, DQ was found to induce autophagy. Exposure of PC12 cells to DQ led to the production of reactive oxygen species (ROS), and N-acetyl-cysteine (NAC) antioxidant effectively blocked both apoptosis and autophagy. Interestingly, DQ in PC12 cells showed increased p53 and NF-κB in a time-dependent manner; furthermore, pifithrin-α (PFT-α), a p53 inhibitor, downregulates the cytotoxicity of DQ, as shown by decreased LC3-II and cleaved caspase-3. SN50, an NF-κB inhibitor, results in diminished LC3-II, cleaved caspase-3, and p53. DQ induces mitogen-activated protein kinase (MAPK) signaling including ERK, JNK, and p38, which inhibit regulated apoptosis and autophagic cell death by controlling mTOR signaling. In addition, modulation of DQ-induced apoptosis in response to autophagy regulation was investigated. Pretreatment with rapamycin, an autophagy inducer, significantly enhanced the viability of DQ-exposed cells by alleviating DQ-induced apoptosis. Conversely, cell pretreatment with 3-methyladenine (3MA), an autophagy inhibitor increased DQ toxicity. Our results suggest that DQ-induced cytotoxicity is modified by autophagy regulation. Pharmacologic induction of autophagy may be a useful treatment strategy in neurodegenerative disorders.
    Keywords:  Apoptosis; Autophagy; Diquat (DQ); MAPKs; NF-κB; P53; mTOR
    DOI:  https://doi.org/10.1007/s00204-019-02424-7
  4. Antioxid Redox Signal. 2019 Mar 02.
      AIMS: Adaptation to low oxygen of hematopoietic stem cells (HSCs) in the bone marrow (BM) has been demonstrated to depend on the activation of hypoxia-inducible factor (HIF)-1α as well as limited production of reactive oxygen species (ROS). In this study, we aimed to determine whether HIF-1α are involved in protecting HSCs from ROS.RESULTS: Oxidative stress was induced by BSO-treatment (DL-buthionine-(S,R)-sulfoximine), which increases the mitochondrial ROS level. While hypoxia rescued Lineage-Sca-1+c-kit+ (LSK) cells from BSO-induced apoptosis, cells succumbed to apoptosis in normoxia. Apoptosis in normoxia was inhibited with the antioxidant N-acetyl-L-cysteine (NAC) or by overexpression of anti-apoptotic BCL-2. Moreover, stabilized expression of oxygen-insensitive HIFs could not protect LSK cells from oxidative stress-induced apoptosis at normoxia, neither could shRNA to HIF-1α inhibit the protective effects by hypoxia in LSK cells. Likewise, BSO treatment of LSK cells from Hif-1 knockout mice did not suppress the effects seen in hypoxia. Microarray analysis identified the nuclear factor kappaB (NF-kB) pathway as a pathway induced by hypoxia. By using NF-kB lentiviral construct and DNA-binding assay we found increased NF-kB activity in cells cultured in hypoxia compared to normoxia. Using an inhibitor against NF-kB activation, we could confirm involvement of NF-kB signaling as BSO-mediated cell death was significantly increased in hypoxia after adding the inhibitor.
    INNOVATION: HIF-1α is not involved in protecting HSCs and progenitors to elevated levels of ROS upon GSH depletion during hypoxic conditions.
    CONCLUSION: The study propose a putative role of NF-kB signaling as a hypoxia-induced regulator in early hematopoietic cells.
    DOI:  https://doi.org/10.1089/ars.2018.7551
  5. Food Chem Toxicol. 2019 Feb 28. pii: S0278-6915(19)30097-3. [Epub ahead of print]
      The present investigation was designed to determine molecular and cellular events involved in anticancer properties of lutein derived from marigold (Tagetes erecta) petals using Human cervical carcinoma (HeLa) cell lines. In vitro experiments demonstrated that lutein at concentration of 10 μM significantly inhibited proliferation of HeLa cells by up to 62.85% after 24 h of treatment and 84.85% after 48 h of treatment. In addition, lutein inhibited proliferation of HeLa cells in a dose-dependent manner by inducing apoptosis. Lutein-treated HeLa cells also showed enhanced accumulation of reactive oxygen species (ROS) correlated with significant downregulation of Bcl-2 (anti-apoptotic) expression and upregulation of Bax (pro-apoptotic) expression. Furthermore, lutein mediated activation of caspase-3 and imbalance between Bax and Bcl-2 expression, causing significant loss of mitochondrial membrane potential of HeLa cells. TUNEL assays revealed significant damage of nuclei DNA in lutein-treated HeLa cells, demonstrating a critical role of lutein in the final stage of apoptosis. Taken together, the results indicate that lutein-induced apoptosis of HeLa cells occurs through enhanced ROS production, interaction with mitochondrial factors, and upregulation of caspase-3-mediated pathway, leading to fragmentation of nuclei DNA. Therefore, lutein could be potentially useful as a chemotherapeutic and/or chemopreventive biomolecule against Human cervical carcinoma.
    Keywords:  Carotenoids; DNA fragmentation; Mitochondrial membrane potential; Oxidative stress-mediated apoptosis; TUNEL assay
    DOI:  https://doi.org/10.1016/j.fct.2019.02.037
  6. Molecules. 2019 Mar 05. pii: E913. [Epub ahead of print]24(5):
      Artemisia absinthium L. has pharmaceutical and medicinal effects such as antimicrobial, antiparasitic, hepatoprotective, and antioxidant activities. Here, we prepared A. absinthium ethanol extract (AAEE) and its subfractions including petroleum ether (AAEE-Pe) and ethyl acetate (AAEE-Ea) and investigated their antitumor effect on human hepatoma BEL-7404 cells and mouse hepatoma H22 cells. The cell viability of hepatoma cells was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The apoptosis, cell cycle, mitochondrial membrane potential (Δψm), and reactive oxygen species (ROS) were analyzed by flow cytometry. The levels of proteins in the cell cycle and apoptotic pathways were detected by Western blot. AAEE, AAEE-Pe, and AAEE-Ea exhibited potent cytotoxicity for both BEL-7404 cells and H22 cells through the induction of cell apoptosis and cell cycle arrest. Moreover, AAEE, AAEE-Pe, and AAEE-Ea significantly reduced Δψm, increased the release of cytochrome c, and promoted the cleavage of caspase-3, caspase-9, and poly(ADP-ribose) polymerase (PARP) in BEL-7404 and H22 cells. AAEE, AAEE-Pe, and AAEE-Ea significantly upregulated the levels of ROS and C/EBP-homologous protein (CHOP). Further, AAEE, AAEE-Pe, and AAEE-Ea significantly inhibited tumor growth in the H22 tumor mouse model and improved the survival of tumor mice without side effects. These results suggest that AAEE, AAEE-Pe, and AAEE-Ea inhibited the growth of hepatoma cells through induction of apoptosis, which might be mediated by the endoplasmic reticulum stress and mitochondrial-dependent pathway.
    Keywords:  Artemisia absinthium; apoptosis; endoplasmic reticulum stress; mitochondrial-dependent pathway
    DOI:  https://doi.org/10.3390/molecules24050913
  7. Folia Morphol (Warsz). 2019 Mar 05.
      Acetaminophen (APAP) hepatotoxicity is characterized by an extensive oxidative stress due to depletion of glutathione (GSH), which results in massive lipid peroxidation and subsequent liver injury. The current paradigm suggests that mitochondria are the main source of reactive oxygen species (ROS), which impair mitochondrial function and are responsible for cell signaling resulting in cell death. This study was designed to compare the potential impact of thymoquinone (THQ), and/or curcumin (CURC) on liver injury induced by APAP toxicity in rats. Serum levels of alanine transaminase (ALT), aspartate transaminase (AST), total bilirubin, and total protein were measured. In addition, liver nitric oxide (NO), malondialdehyde (MDA), reduced glutathione (GSH), and superoxide dismutase (SOD) were estimated. Moreover, these biochemical parameters were confirmed by histopathological and immunohistochemical investigations for the expression of thioredoxin, iNOS and caspase 3. Acetaminophen toxicity elevated most of the above-mentioned parameters but decreased GSH, SOD, and total protein levels. Histologically, liver sections demonstrated liver injury characterized by hepatocellular necrosis with nuclear pyknosis, karyorrhexis and karyolysis. Immunohistochemical study revealed increased expression of iNOS and caspase 3 proteins, while the thioredoxin protein expression was decreased. Treatment with the THQ and CURC regulated the biochemical and histopathological alterations induced by APAP toxicity. It was concluded that the combination strategy of THQ and CURC might be considered as a potential antidote in combating liver injury induced by APAP with minimal side effects.
    Keywords:  acetaminophen; curcumin; hepatotoxicity; thymoquinone
    DOI:  https://doi.org/10.5603/FM.a2019.0027
  8. Bioorg Med Chem. 2019 Mar 02. pii: S0968-0896(18)31939-4. [Epub ahead of print]
      The natural compound 1,4-naphthoquinone has potent anti-tumor activity. However, the clinical application of 1,4-naphthoquinone and its derivatives has been limited by their side effects. In this study, we attempted to reduce the toxicity of 1,4-naphthoquinone by synthesizing two derivatives: 2,3-dihydro-2,3-epoxy-2-propylsulfonyl-5,8-dimethoxy-1,4-naphthoquinone (EPDMNQ) and 2,3-dihydro-2,3-epoxy-2-nonylsulfonyl-5,8-dimethoxy-1,4-naphthoquinone (ENDMNQ). Then we evaluated the cytotoxicity and molecular mechanisms of these compounds in lung cancer cells. EPDMNQ and ENDMNQ significantly inhibited the viabilities of three lung cancer cell lines and induced A549 cell cycle arrest at the G1 phase. In addition, they induced the apoptosis of A549 lung cancer cells by increasing the phosphorylation of p38 and c-Jun N-terminal kinase (p-JNK), and decreasing the phosphorylation of extracellular signal-related kinase (p-ERK), protein kinase B (Akt), and signal transducer and activator of transcription 3 (STAT3). Furthermore, they increased reactive oxygen species (ROS) levels in A549 cells; however, pretreatment with the ROS inhibitor N-acetyl-l-cysteine significantly inhibited EPDMNQ- and ENDMNQ-mediated apoptosis and reversed apoptotic proteins expression. In conclusion, EPDMNQ and ENDMNQ induced G1 phase cell cycle arrest and apoptosis in A549 cells via the ROS-mediated activation of mitogen activated protein kinase (MAPK), Akt and STAT3 signaling pathways.
    Keywords:  1,4-Naphthoquinone; Akt; Apoptosis; Lung cancer; MAPK; ROS; STAT3
    DOI:  https://doi.org/10.1016/j.bmc.2019.03.002
  9. Sci Rep. 2019 Mar 08. 9(1): 3969
      The Warburg effect, wherein cancer cells prefer glycolysis rather than oxidative phosphorylation even under normoxic conditions, is a major characteristic of malignant tumors. Lactate dehydrogenase A (LDHA) is the main enzyme regulating the Warburg effect, and is thus, a major target for novel anti-cancer drug development. Through our ongoing screening of novel inhibitors, we found that several selenobenzene compounds have inhibitory effects on LDHA activity. Among them, 1-(phenylseleno)-4-(trifluoromethyl) benzene (PSTMB) had the most potent inhibitory effect on the enzymatic activity of LDHA. The results from biochemical assays and computational modeling showed that PSTMB inhibited LDHA activity. In addition, PSTMB inhibited the growth of several tumor cell lines, including NCI-H460, MCF-7, Hep3B, A375, HT29, and LLC. In HT29 human colon cancer cells, PSTMB dose-dependently inhibited the viability of the cells and activity of LDHA, without affecting the expression of LDHA. Under both normoxic and hypoxic conditions, PSTMB effectively reduced LDHA activity and lactate production. Furthermore, PSTMB induced mitochondria-mediated apoptosis of HT29 cells via production of reactive oxygen species. These results suggest that PSTMB may be a novel candidate for development of anti-cancer drugs by targeting cancer metabolism.
    DOI:  https://doi.org/10.1038/s41598-019-40617-3
  10. Eur Rev Med Pharmacol Sci. 2019 Feb;pii: 17143. [Epub ahead of print]23(4): 1797-1806
      OBJECTIVE: The aim of this study was to explore the protective effect of nicotinamide on hypoxic cardiomyocytes and to investigate its possible mechanism.MATERIALS AND METHODS: Primary cardiomyocytes were used as study subjects. They were divided into three groups, including the blank group, control group and nicotinamide pretreatment group. Cell counting kit-8 (CCK-8) was used to detect cell viability. Lactate dehydrogenase (LDH) was used to detect cytotoxicity and flow cytometry was used to detect cell apoptosis. Polymerase Chain Reaction (PCR) and Western blot were used to measure the expressions of genes in adenosine monophosphate-activated protein kinase (AMPK) pathway. JC-1 detected the levels of mitochondrial membrane potential and reactive oxygen species (ROS). NAD was used for nicotinamide adenine dinucleotide (NAD)+, and NAD phosphate (NADP)+ levels. Adenosine triphosphate (ATP) assay was performed for the detection of intracellular energy metabolism.
    RESULTS: In the absence of oxygen, nicotinamide had a protective effect on primary cardiomyocytes. Meanwhile, nicotinamide could markedly inhibit the increase of caspase3 mRNA in cardiomyocyte apoptosis pathway, and suppress the expression of apoptotic proteins. Furthermore, it could significantly induce the increase of intracellular ATP and activate the AMPK pathway. The detection of mitochondria indicated that nicotinamide alleviated hypoxic cardiomyocytes. In addition, the mitochondrial membrane potential disrupted and inhibited mitochondrial oxidative stress levels.
    CONCLUSIONS: Nicotinamide pretreatment protects hypoxic cardiomyocytes and reduces intracellular mitochondrial stress. This protection may be related to the induction of the AMPK pathway and the increase of intracellular energy production.
    DOI:  https://doi.org/10.26355/eurrev_201902_17143
  11. Reprod Biol. 2019 Feb 28. pii: S1642-431X(18)30195-5. [Epub ahead of print]
      Doxorubicin is an extensively applied anti-cancerous drug since 1950's and its usage is constrained because of its accumulation in a non-cancerous organ. Many studies have proven that doxorubicin causes reproductive toxicity depends on its dosage, particularly due to increased oxidative stress and apoptosis. A number of the researches have been carried out concerning its prevention. But there is a need to recognize the mechanism at the back of its toxicity to get better and improved method of treatment. To clarify the feasible mechanism of doxorubicin-mediated reproductive toxicity in rats, we have administrated doxorubicin at distinct dosages inclusive of low dosage (male rats that are at 230-250 g acquired cumulatively 1.5 mg/kg; ip; once per week for five weeks) and high dosage (male rats which are at 230-250 grams obtained cumulatively 15 mg/kg; ip; once every week for five weeks). Doxorubicin decreases antioxidant level such as GSH, Cu/Zn SOD, Mn SOD both in serum and testes. Increased oxidative stress is considered via elevated MDA level both in serum and testes. The level of ROS is measured via the DCFDA method in testes. Apoptosis become found through DNA fragmentation assay and quantification of Caspase 3, Caspase 9, Bcl2 and Cytochrome C. Doxorubicin mediated oxidative stress and apoptosis in testicular milieu is through deregulation of Nrf2, PGC-1α, AHR, ARNT, PXR, SUMO-1, UCP2, UCP3, ANX A5, Caspase 3, Caspase 9, Bcl2, Cytochrome C, GR, and GPX. In end, doxorubicin-mediated oxidative stress and apoptosis is through diverse transcriptional factors and genes with respect to decreased antioxidant level, augmented ROS level and Annexin A5 in the testicular milieu.
    Keywords:  Antioxidants; Apoptosis; Doxorubicin; Male reproductive toxicity; Oxidative stress; Transcription factors
    DOI:  https://doi.org/10.1016/j.repbio.2019.02.004
  12. Biomed Pharmacother. 2019 Mar;pii: S0753-3322(18)37012-4. [Epub ahead of print]111 1315-1325
      Diabetic retinopathy (DR) seriously endangers human beings' health, uncovering the underlying mechanism might help to cure DR. In this study, we found that the effects of glucose on retinal pigment epithelium (RPE) varies in a dose dependent manner, high-glucose (50mM) promotes reactive oxygen species (ROS) generation and cell apoptosis, inhibits cell mitophagy as well as proliferative abilities, while low-glucose (15mM) induces ROS production and cell mitophagy, but has little impacts on cell apoptosis and proliferation. Of note, the toxic effects of high-glucose (50mM) on RPE are alleviated by ROS scavengers and aggravated by autophagy inhibitor 3-methyladenine (3-MA) or mitophagy inhibitor cyclosporin A (CsA). High-glucose (50mM) induced ROS generation is merely eliminated by ROS scavengers instead of mitophagy or autophagy inhibitor. We also proved that high-glucose (50mM) inhibits cell proliferation and promotes cell apoptosis by regulating ROS mediated inhibition of mitophagy. In addition, mitophagy associated proteins PINK1 and Parkin are downregulated by high-glucose (50mM) or hydrogen peroxide treatments, which are reversed by ROS scavengers. Of note, Knock-down of PINK1 decreases phospharylated Parkin instead of total Parkin levels in RPE. Intriguingly, high-glucose's inhibiting effects on cell mitophagy as well as proliferation and its promoting effects on cell apoptosis are reversed by either PINK1 or Parkin overexpression. Therefore, we concluded that high-glucose promotes RPE apoptosis and inhibits cell proliferation as well as mitophagy by regulating ROS mediated inactivation of ROS/PINK1/Parkin signal pathway.
    Keywords:  Apoptosis; Diabetic retinopathy; High-glucose; Mitophagy; RPE
    DOI:  https://doi.org/10.1016/j.biopha.2019.01.034
  13. Photodiagnosis Photodyn Ther. 2019 Mar 02. pii: S1572-1000(18)30402-2. [Epub ahead of print]
      OBJECTIVES: The aim of this study was to develop a novel photosensitizer from traditional plant extracts and to investigate the photodynamic therapy (PDT) effect and mechanism of action of the novel photosensitizer on KB and Hep-2 cells.METHODS: Fluorescence emission, cell viability, and intracellular distribution of candidates were analyzed to screen potential photosensitizers from traditional plant extracts. Cellular reactive oxygen species (ROS) quantification, Annexin V-FITC/PI staining, and western blotting were performed to explore the mechanism of cell death in KB and Hep-2 cells.
    RESULT: Of 289 traditional plant extracts, 13 plant extracts with strong fluorescence were initially screened by fluorescence emission analysis. The cell viability assay and intracellular distribution of candidates showed that Acanthopanacis Cortex (AC) extract is a potential photosensitizer. Under optimal PDT conditions, high levels of ROS were produced in KB and Hep-2 cells, followed by cell death. However, there was no significant damage to HaCaT cells. Moreover, apoptosis induced by AC extract with 625 nm irradiation (IR) down-regulated the expression of Bcl-2 protein and up-regulated the expression of Bax protein, as well as that of cleaved PARP-1 protein in both KB and Hep-2 cells.
    CONCLUSION: The fluorescence intensity of AC extract at 420 nm is similar to that of the commercial Hematoporphyrin (HP). AC extract with 625 nm IR could enhance the PDT effect, induce ROS generation, and trigger apoptotic pathways in KB and Hep-2 cells. Therefore, we suggest that AC is a potential novel photosensitizer for PDT in head and neck squamous cell carcinoma.
    Keywords:  Acanthopanacis Cortex; Apoptosis; Head and neck squamous cell carcinoma; Natural photosensitizer; Photodynamic therapy
    DOI:  https://doi.org/10.1016/j.pdpdt.2019.02.020
  14. Ecotoxicol Environ Saf. 2019 Mar 04. pii: S0147-6513(19)30226-X. [Epub ahead of print]174 270-282
      Triclosan (TCS) is an antimicrobial preservative used in personal care products. Here, we have studied the phototoxicity, photogenotoxicity of TCS and its molecular mechanism involving p38 mitogen activated protein kinase (MAPK) pathway under UVB/sunlight exposure. We found that TCS showed photodegradation and photoproducts formation under UVB/sunlight. In silico study suggests that photosensitized TCS loses its preservative property due to the formation of its photoproducts. Photosensitized TCS induces significant O2•-, •OH generation and lipid peroxidation via type-I photochemical reaction mechanism under UVB/sunlight exposure. We performed intracellular study of TCS on human skin keratinocytes (HaCaT cell-line) under the ambient intensity of UVB (0.6 mW/cm2) and sunlight exposure. Significant intracellular ROS generation was observed through DCFH2-DA/DHE assays along with a significant reduction in cell viability through MTT and NRU assays in photosensitized TCS. Photosensitized TCS also induces endoplasmic reticulum (ER) stress as shown through ER-tracker/DAPI staining and Ca2+ release. It further induced cell cycle arrest through the sub-G1 phase augmentation and caused lysosomal/mitochondrial destabilization. Photogenotoxicity was shown through significant tail DNA, micronuclei and cyclobutane pyrimidine dimers (CPDs) formations. Cell signaling mechanism implicated upregulated expression of cleaved Caspase-3, Bax, phospho-p38, phospho-JNK and cytochrome C, thereby downregulated Bcl-2 expressions. Results advocate that TCS induces phototoxic effects via type I mediated photodynamic mechanism and activation of MAPK pathway. We conclude that photoexcited TCS may be deleterious to human health at the ambient environmental intensities of sunlight reaching at the earth's surface. Therefore, it may be replaced by alternative safe preservative.
    Keywords:  Apoptosis; DNA damage; HaCaT; Reactive oxygen species; Triclosan; p38
    DOI:  https://doi.org/10.1016/j.ecoenv.2019.02.065
  15. Future Oncol. 2019 Mar 07.
      AIM: Curcumin induces cytotoxic cell death in several human cancer cells. Here, we have investigated the effects of curcumin on non-small-cell lung cancer (NSCLC) with an aim to identify underlying mechanisms of its cytotoxic effect.MATERIALS & METHODS: The effects of various concentrations of curcumin on the NSCLC cell lines A549 and SPC-A1 were evaluated by MTT assay, colony-forming assay and flow cytometry. Additionally, protein expression associated with different signaling pathways was assessed using western blotting.
    RESULTS: Curcumin exhibited cytotoxicity against NSCLC, evident from the inhibition of cell proliferation, G2/M arrest, DNA damage, endoplasmic reticulum stress and mitochondrial apoptosis. The anticancer effect was related to reactive oxygen species (ROS) accumulation and could be reversed by ROS scavengers, catalase and N-acetyl-l-cysteine. Curcumin decreased mitochondrial transmembrane potential and induced ROS production, thereby activating the DNA damage/repair pathway and mitochondrial apoptosis.
    CONCLUSION: These results indicate that curcumin could be an effective therapeutic candidate for NSCLC.
    Keywords:  DNA damage; ER stress; curcumin; mitochondrial apoptosis; non-small-cell lung cancer; reactive oxygen species
    DOI:  https://doi.org/10.2217/fon-2018-0708
  16. Eur Rev Med Pharmacol Sci. 2019 Feb;pii: 17134. [Epub ahead of print]23(4): 1722-1729
      OBJECTIVE: Phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling pathway plays an important role in regulating cell survival, apoptosis and oxidative stress (OS). Phosphatase and tensin homolog deleted on chromosome ten (PTEN) can negatively regulate PI3K/AKT signaling pathway. DJ-1 is also a key negative regulator of PTEN. DJ-1-PTEN/PI3K/AKT signaling pathway regulates ischemia reperfusion (I-R). This study investigated the role of DJ-1 in affecting myocardial I-R injury.MATERIALS AND METHODS: The rat myocardial I-R injury model was established. Expression of DJ-1 and PTEN in myocardial tissue was detected. The reactive oxidative species (ROS) content was detected using flow cytometry. Caspase-3 activity, malondialdehyde (MDA) content, and superoxide dismutase (SOD) activities were determined by ultraviolet spectrophotometry. Rat cardiomyocytes H9C2 were cultured in vitro and divided into control group, I-R group, I-R+pIRES2-NC group, and I-R+pIRES2-DJ-1 group. Levels of DJ-1, PTEN and phosphorylated AKT (p-AKT) were detected. Cell apoptosis and ROS content were evaluated using flow cytometry.
    RESULTS: Compared with sham group, caspase-3 activity, MDA content, and PTEN expression were significantly increased, while SOD activity and DJ-1 levels were significantly reduced in myocardial tissue of I-R group (p<0.05). Compared with the control, I-R treatment markedly induced H9C2 cell apoptosis, decreased DJ-1 and p-AKT expression, and enhanced ROS production and PTEN expression. DJ-1 overexpression apparently down-regulated PTEN expression, elevated p-AKT level, and attenuated apoptosis and ROS production in H9C2 cells (p<0.05).
    CONCLUSIONS: Abnormal expression of DJ-1 plays a regulatory role in the process of myocardial I-R injury. Over-expression of DJ-1 can reduce myocardial cell I-R damage sensitivity by inhibiting PTEN expression, enhancing the activity of PI3K/AKT signaling pathway, reducing ROS production, and alleviating apoptosis.
    DOI:  https://doi.org/10.26355/eurrev_201902_17134
  17. J Leukoc Biol. 2019 Mar 05.
      We studied the effects of a synthetic CpG oligonucleotide (CpG ODN2006) on polymorphonuclear leukocyte (PMNL, neutrophil) survival and oxidant status. CpG ODN2006 showed a dose-dependent effect on the apoptosis of resting neutrophils. Without affecting the viability of resting cells, low concentrations of CpG ODN2006 interfered with Salmonella typhimurium-mediated viability prolongation and increased neutrophil apoptosis to control levels. CpG ODN2006 stimulated neutrophil apoptosis by enhancing ROS generation. Even small doses of ODN could induce the production of intracellular superoxide anions. The high superoxide reactogenicity, including with respect to nitrogen oxide, led to increased levels of intracellular ROS and RNS, which ultimately caused apoptosis. The pro-oxidant effect of low concentrations of CpG ODN2006 was not sufficient to trigger irreversible pro-apoptotic mechanisms. However, the sensitivity of PMNLs to ODN2006, a modulator of apoptosis, increased significantly under conditions of infectious inflammation. Inactivated S. typhimurium proved to be suitable for simulating inflammatory conditions in vitro.
    Keywords:  CpG oligonucleotide; Salmonella; apoptosis; neutrophil; reactive nitrogen species; reactive oxygen species
    DOI:  https://doi.org/10.1002/JLB.3MIA1118-435R
  18. Cancer Sci. 2019 Mar 06.
      Inducing angiogenesis is a hallmark of cancers that sustains tumor growth and metastasis. Neovascularization is a surprisingly early event during the multistage of cancer progression. Cinobufagin, an important bufadienolide originates from Chan'Su, has been clinically used to treat cancer in China since Tang dynasty. Here we show that cinobufagin suppresses colorectal cancer (CRC) growth in vivo by downregulating angiogenesis. The hierarchized neovasculature is significantly decreased and the vascular network formation is disrupted in human umbilical vein endothelial cells (HUVECs) by cinobufagin in a dose-dependent manner. Endothelial apoptosis is observed by inducing reactive oxygen species (ROS) accumulation and mitochondrial dysfunction which can be neutralized by N-acetyl-L-cysteine (NAC). The expression of hypoxia-inducible factor 1α (HIF-1α) is reduced and phosphorylation of mechanistic target of rapamycin (mTOR) at Ser2481 and Akt at Ser473 is downregulated in HUVECs. The endothelial apoptosis is triggered by cinobufagin via stimulation of Bax and cascade activation of caspase 9 and caspase 3. Increased endothelial apoptosis rate and alterations in HIF-1α/mTOR pathway are recapitulated in tumor bearing mice in vivo. Further, the anti-angiogenesis function of cinobufagin is consolidated based on its pro-apoptotic effects on an EOMA-derived hemangioendothelioma model. In conclusion, cinobufagin suppresses tumor neovascularization by disrupting endothelial mTOR/HIF-1α pathway to trigger ROS-mediated vascular endothelial cell apoptosis. Cinobufagin is a promising natural anti-angiogenetic drug, which has clinical translation potential and practical application value. This article is protected by copyright. All rights reserved.
    Keywords:  Angiogenesis; Apoptosis; Cinobufagin; Colorectal cancer; HIF-1α
    DOI:  https://doi.org/10.1111/cas.13988
  19. Redox Biol. 2019 Mar 02. pii: S2213-2317(18)31230-8. [Epub ahead of print]22 101157
      Ultraviolet B (UVB) irradiation can induce reactive oxygen species (ROS) production and apoptosis in human lens epithelial cells (HLECs), thus leading to the formation of cataracts. We studied the role of tripartite motif 69 (TRIM69) in cataract formation. The expression of TRIM69 protein was down-regulated in both human cataract capsule tissues and HLECs treated with UVB, whereas the expression of p53 protein exhibited an opposite trend. Ectopic expression of TRIM69 in HLECs significantly suppressed UVB-induced apoptosis and ROS production, whereas knockdown of TRIM69 promoted apoptosis and ROS production. TRIM69 can interact with p53 and induce its ubiquitination. The effects of TRIM69 overexpression in UVB-induced cell apoptosis and ROS production was clearly weakened by p53 overexpression, thus suggesting a role for p53 in TRIM69 functions. Furthermore, inhibition of ROS mitigated the effects of UVB irradiation on ROS production, cell apoptosis, forkhead box protein 3a (Foxo3a) phosphorylation, and TRIM69 expression. Additionally, Foxo3a overexpression significantly enhanced TRIM69 promoter activity, whereas Foxo3a knockdown had the opposite effect. In conclusion, we provide the first demonstration that Foxo3a is a potential transcription factor for TRIM69, and TRIM69 induces p53 ubiquitination. These results suggest that the Foxo3a/TRIM69/p53 regulatory network may be involved in cataract formation.
    Keywords:  Cataract; Foxo3a; TRIM69; UVB; p53
    DOI:  https://doi.org/10.1016/j.redox.2019.101157
  20. Pharmacology. 2019 Mar 06. 103(5-6): 303-314
      BACKGROUND: Focal and segmental glomerular sclerosis (FSGS) is a common cause of nephrotic syndrome and end-stage renal disease. It has been reported that overproduction of reactive oxygen species (ROS) and cell apoptosis are associated with the development of FSGS. Epigallocatechin-3-gallate (EGCG) is a bioactive constituent accounting for more than 50% of the total catechins in green tea, which have anti-oxidative and anti-apoptotic effects. Based on this, this study was designed to evaluate the renoprotective effect of EGCG treatment on Adriamycin-induced FSGS. -Methods: In C57BL/6 mice, Adriamycin nephropathy (AN) was induced by Adriamycin (10 mg/kg body weight, diluted in normal saline) via a tail vein on day 0. Then the mice were given with EGCG (20 mg/kg body weight) or YC-1 (Lificiguat, a specific inhibitor of hypoxia-inducible factor-1α [HIF-1α], 50 mg/kg body weight) or both intraperitoneally. Both the EGCG and YC-1 were given on the day of Adriamycin injection and continued for 6 weeks. The animals were organized into the following 5 groups for the animal experiments: the control group, the AN group, the AN + EGCG group, the AN + YC-1 group and the AN + EGCG + YC-1 group. At 6 weeks, the mice were sacrificed; kidneys and blood samples were collected for further analysis. The HIF-1α and the angiopoietin-like 4 (ANGPTL4) expression were detected by Western blot, real-time PCR, immunohistochemistry or immunofluorescence. Dihydroethidium staining and NADPH oxidase 1 (Nox1) measurement were used to detect ROS production. Terminal deoxynucleotide transferase-mediated dUTP nick end-labeling (TUNEL) staining and caspase-3 measurement was used to detect cell apoptosis.RESULTS: When the animals were treated with Adriamycin, both the ROS production and TUNEL positive cells increased. Besides, the expression of HIF-1α, ANGPTL4, and caspase-3 were also up-regulated, while EGCG treatment could attenuate these changes. Interestingly, compared with treatment with YC-1 or EGCG alone, more pronounced inhibition of ANGPTL4, caspase-3 and Nox1 were obtained when YC-1 and EGCG were administered simultaneously.
    CONCLUSION: EGCG attenuates FSGS through the suppression of Oxidant Stress and apoptosis by targeting the HIF-1α/ANGPTL4 pathway.
    Keywords:  Adriamycin nephropathy; Angiopoietin-like 4; Apoptosis; Epigallocatechin-3-gallate; Focal segmental glomerulosclerosis; Hypoxia-inducible factor-1α; Oxidant stress
    DOI:  https://doi.org/10.1159/000496799
  21. Eur Rev Med Pharmacol Sci. 2019 Feb;pii: 17135. [Epub ahead of print]23(4): 1730-1741
      OBJECTIVE: Nuclear factor NF-E2 related factor 2 (Nrf2) plays crucial roles in the regulation of oxidative stress (OS) or myocardial ischemia-reperfusion (I-R) injury. During the process of I-R injury, the miR-320 is down-regulated. Bioinformatics analysis showed complementary binding sites between miR-320 and 3'-UTR of Nrf2 mRNA. Therefore, this study aimed to investigate the role of miR-320 in mediating Nrf2 expression and myocardial I-R injury.MATERIALS AND METHODS: Rat I-R model was established. Fluorescent quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) and Western blot were used to measure the expression of miR-320, Nrf2, HO-1 in myocardial tissues. Contents of malondialdehyde (MDA), superoxide dismutase (SOD) and caspase-3 activity, serum assay of creatine kinase (CK) and lactate dehydrogenase (LDH) activity were evaluated. I-R rat models were transfected with antagomir-320 followed by measuring those proteins. Cultured H9C2 cells were transfected with antagomir-320 to measure miR-320, Nrf2 and heme oxygenase 1 (HO-1) expression, cell apoptosis and reactive oxygen species (ROS) content.
    RESULTS: Compared to the sham group, I-R rats had significantly lower miR-320 or HO-1 expression in the myocardium, plus higher levels of Nrf2, MDA, CK and LDH, and decreased SOD activity (p<0.05). Antagomir-320 transfection suppressed miR-320 expression, elevated Nrf2 and HO-1 expression, decreased levels of MDA, CK or LDH, and increased SOD activity. In H9C2 cells, antagomir-320 transfection also elevated Nrf2 and HO-1 expression and suppressed myocardial cell apoptosis or ROS production under I-R treatment.
    CONCLUSIONS: Down-regulation of miR-320 exerts protective effects on myocardial I-R injury. The inhibition of mir-320 expression can enhance OS potency of the myocardium, alleviate I-R injury or reduce cell apoptosis by facilitating Nrf2 expression.
    DOI:  https://doi.org/10.26355/eurrev_201902_17135
  22. Phytother Res. 2019 Mar 05.
      In this study, the antitumor activity of two furanoflavanoid derivatives, Pongapin and Karanjin, was evaluated in comparison with Plumbagin, a plant-derived polyphenol with proven antitumor activity. The compounds differentially inhibit the growth of different cancer cell lines (most effective on HeLa cells), with very low inhibitory effect on the growth of normal mouse embryonic fibroblast cell line. Pongapin like Plumbagin could significantly increase the intracellular reactive oxygen species (ROS) in the HeLa cells by stabilization of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor (I-κB) expression and reduction of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) expression. In contrast, Karanjin could decrease ROS level by inhibition of I-κB degradation resulting restriction of NF-κB nuclear translocation. Pongapin and Plumbagin significantly increased DNA damage-induced p53 expression and p21 nuclear expression. However, Karanjin treatment showed low DNA damage with increased p53 expression. The compounds induced G2/M arrest and increase in SubG1 population, indicating induction of apoptosis. Apoptosis was further validated by acridine orange/ethidium bromide dual staining and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay in HeLa cells after treatment with the compounds. The compounds induced caspase-dependent apoptosis through induction of Bax/Bcl-2 ratio either through increased expression of Bax by Pongapin and Plumbagin or low expression of Bcl-2 by Karanjin. Thus, Pongapin and Karanjin may be potential natural anticancer agents in the future, like Plumbagin.
    Keywords:  Apoptosis; Karanjin; NF-κB; Plumbagin; Pongapin; reactive oxygen species
    DOI:  https://doi.org/10.1002/ptr.6302
  23. Iran J Basic Med Sci. 2019 Feb;22(2): 154-159
      Objectives: The main goal of the current research was to examine the effects of Berberine (BBR) on apoptotic signaling and hippocampal oxidative stress induced by common carotid artery occlusion.Materials and Methods: Chronic cerebral hypoperfusion (CCH) model was created by occluding the two common carotid arteries (two-vessel occlusion [2VO]) permanently. BBR (50 and 100 mg/kg/daily) was intra-gastrically administered to ischemic rats. Neuronal survival was evaluated by Nissl staining. The levels of malondialdehyde (MDA) and antioxidant enzymes, including catalase (CAT) and superoxide dismutase (SOD), along with the activities of caspase 3 were estimated in the hippocampus 2 month after treating the rats with 2VO.
    Results: According to findings of the present research, the BBR therapy inhibited the neuro-degeneration of hippocampus. BBR also significantly decreased the amount of MDA and activity of caspase 3 in the hippocampus. Furthermore, the administration of BBR alleviated the lowered activities of SOD and CAT after 2VO surgery.
    Conclusion: The antioxidant and antiapoptotic properties of BBR might play important roles in improving functional outcomes and might have significant neuroprotective effects on the CCH damage.
    Keywords:  Antioxidant enzymes; Apoptosis; Berberine; Chronic cerebral – hypoperfusion; Common carotid artery MDA; Rat
    DOI:  https://doi.org/10.22038/ijbms.2018.31225.7534
  24. Food Chem Toxicol. 2019 Mar 04. pii: S0278-6915(19)30102-4. [Epub ahead of print]
      Hesperidin is a flavonoid which occurs in citrus fruits. Hesperidin was gamma-irradiated at doses of 0, 30, 70, and 150 kGy. Gamma irradiation induced a decreased hesperidin peak, and a new radiolytic peak that gradually increased up to 150 kGy. The new radiolytic peak was fractionated, and the fractionated hesperidin derivative was used for subsequent experiments. Hesperidin gamma-irradiated at 150 kGy was toxic toward B16BL6 cells, but not toward bone marrow-derived macrophages. This cytotoxicity was exerted via induction of apoptosis, as reflected by the high population of double-positive cells, increased sub-G1 phase cells, depolarization of matrix metalloproteinase, production of reactive oxygen species, weakness of cell adhesion, changes in cell morphology, and inhibition of B16BL6 cell migration. Furthermore, 150 kGy gamma-irradiated hesperidin decreased the expression of Bcl-2 and pro-caspases-3 and -9, increased the expression of Bax and cytosolic cytochrome c, and increased the cleavage of poly ADP ribose polymerase. In vitro mechanistic study revealed that 150 kGy gamma-irradiated hesperidin achieved significantly greater inhibition of lung metastasis and growth of melanoma B16BL6 cells in C57BL/6 mice than non-irradiated intact hesperidin did. These results suggest that the structural modification of hesperidin induced by gamma irradiation could facilitate the development of anti-cancer drugs.
    Keywords:  Apoptosis; Gamma irradiation; Hesperidin; Metastasis; Tumor growth
    DOI:  https://doi.org/10.1016/j.fct.2019.02.042
  25. J Cell Physiol. 2019 Mar 07.
      Chalcones found in fruits and vegetables have promising cancer chemopreventive properties. This study attempts to identify the anticancer efficacies of chalcone flavokawain B (FKB) in the rhizomes of Alpinia pricei Hayata by examining key molecular events in non-small-cell lung cancer (A549) cells. Our results indicated that in human A549 cells, FKB (0-15 μg/ml) decreases cell viability and colony formation, dysregulates the Bax:B-cell lymphoma 2 ratio and increases apoptotic DNA fragmentation. Mitochondrial (caspase-9/-3 and poly ADP ribose polymerase [PARP]) signaling was found to be involved in FKB-induced apoptosis. In addition, FKB-induced reactive oxygen species (ROS) generation, and N-acetylcysteine attenuated FKB-induced apoptotic cell death. Moreover, FKB triggered autophagy, as evidenced by the improved acidic vesicular organelle formation, lipidated light chain 3 (microtubule-related light chain 3) accumulation, and ATG7 expression and the decreased mammalian target of rapamycin phosphorylation. Furthermore, FKB suppressed ROS-mediated ATG4B expression. Inhibiting autophagy using 3-methyladenine/chloroquine diminished FKB-induced cell death, indicating that autophagy is triggered as a death mechanism by FKB. In summary, FKB has a crucial role in the execution and propagation of ROS-mediated apoptotic and autophagic cell death of lung adenocarcinoma cells.
    Keywords:  Alpinia pricei hayata; ROS; apoptosis; autophagy; chalcone flavokawain B
    DOI:  https://doi.org/10.1002/jcp.28375
  26. Pak J Pharm Sci. 2019 Jan;32(1(Supplementary)): 327-331
      Diabetic cardiomyopathy (DC) is a serious complication of diabetes. Apoptosis, inflammatory and ROS production are among the factors that are involved in the progression of diabetic cardiomyopathy. 6-shogaol is reported to inhibit apoptosis and reduce inflammatory and ROS production. This study aimed to study the effect of 6-shogaol (6S) on the progression of diabetic cardiomyopathy in vitro. To develop DC model, H9c2 cell line was exposed to high glucose (HG) level (33 M glucose) for 24 h and used as a model for diabetic cardiomyopathy. Another set of H9c2 cell lines were 1 h pretreated with different conc. of 6-shogaol (5-20 μM). Cell viability, apoptosis, ROS production, IL-6, TNF-alpha and NF-κB were estimated in these cell lines treated with HG level or pretreated with 6-shgoal before HG. Exposing cardiomyocytes H9c2 cells to HG produced dramatic changes in cell biology and chemistry. There is a significant reduction in cell viability and enhancement in cell apoptosis as compared with control. In addition, ROS production, IL-6, TNF-α levels were increased in H9c2 line treated with HG. Also, there is overexpression of NF-κB in cells treated with HG levels alone. On the other hand, pretreatment of cardiomyocytes H9c2 cells with 6-shogaol (5-20μM) significantly improved cell viability and reduced apoptosis, in addition, 6S at a dose of 10 μM abrogated the deleterious effects of HG on oxidative stress and inflammatory parameters via modulation of NF-κB pathway. Therefore, 6S has a potential protective effect against hyperglycemia-induced DC in vitro.
  27. Iran J Basic Med Sci. 2019 Feb;22(2): 140-145
      Objectives: The aim of this study is to explore the potential neuroprotective effects of Ginkgolide B (GB), a main terpene lactone and active component in Ginkgo biloba, in hypoxia-induced neuronal damage, and to further investigate its possible mechanisms.Materials and Methods: 54 Sprague-Dawley rats were randomly divided into three groups: the untreated control group (n=18); the hypoxia group (n=18; exposed to 6000 m simulated plateau altitude for six days); and the GB group (n=18; intragastric administration of 12 mg/kg GB three days prior to rapid adaption to 6000 m and on the first two days of hypoxia). After hypoxia exposure for six days, we dissected out the brain hippocampi and performed hematoxylin and eosin staining, Nissl staining, and TUNEL staining. Homogenates of the hippocampi were used to test the oxidative stress indices including malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), and catalase. Bax and caspase-3 expression in the hippocampal tissue was measured using qRT-PCR.
    Results: Treatment with GB before exposure to hypoxia could protect neural cells and increase the number of Nissl bodies. TUNEL and qRT-PCR results demonstrated that GB treatment could decrease apoptotic cells in different areas of the hippocampus. Antioxidant defense systems such as SOD, GSH, and catalase were decreased (P<0.05), and the concentration of MDA was reduced significantly in the hippocampi of rats of the GB group (P<0.05).
    Conclusion: GB could alleviate hypoxia-induced neuronal damage in rat hippocampus by inhibiting oxidative stress and apoptosis.
    Keywords:  Apoptosis; Ginkgolide B; Hippocampus; Hypoxia; Oxidative stress injury
    DOI:  https://doi.org/10.22038/ijbms.2018.26228.6569
  28. Am J Chin Med. 2019 Mar 04. 1-15
      This study examined whether serotonin and two of its derivatives, N -feruloylserotonin and N -( p -coumaroyl) serotonin, have a renoprotective effect in a mouse model of cisplatin-induced acute renal failure. Cisplatin (20 mg/kg body weight) was administered by intraperitoneal injection to male BALB/c mice that had received oral serotonin, N -feruloylserotonin or N -( p -coumaroyl) serotonin (7.5 mg/kg body weight per day) during the preceding 2 days. At 3 days after the cisplatin injection, serum and renal biochemical factors, oxidative stress, inflammation and apoptosis-related protein expression were evaluated, and histological examinations were performed. Cisplatin caused reduction in body weight and an increase in kidney weight; however, N -( p -coumaroyl) serotonin and N -feruloylserotonin attenuated these effects. Moreover, the serotonin derivatives significantly decreased serum urea nitrogen and creatinine levels. They also significantly reduced the level of reactive oxygen species and upregulated the expression of glutathione peroxidase in the kidney. Furthermore, the serotonin derivatives improved the abnormal expression of mitogen-activated protein kinases activation-dependent inflammation- and apoptosis-related protein and caused less renal damage. These results provide important evidence that N -( p -coumaroyl) serotonin and N -feruloylserotonin exert a pleiotropic effect on several parameters related to oxidative stress, inflammation and apoptosis. The derivatives also have a renoprotective effect in cisplatin-treated mice; however, this effect is higher with N -( p -coumaroyl) serotonin.
    Keywords:  Cisplatin; N-(p-Coumaroyl) Serotonin; N-Feruloylserotonin; Nephrotoxicity; Safflower Seed; Serotonin
    DOI:  https://doi.org/10.1142/S0192415X19500186
  29. J Pharmacol Sci. 2019 Feb 10. pii: S1347-8613(19)30015-5. [Epub ahead of print]
      Ischemia/reperfusion (I/R)-induced oxidative stress is a serious clinical problem in the reperfusion therapy for ischemic diseases. Tumstatin is an endogenous bioactive peptide cleaved from type IV collagen α3 chain. We previously reported that T3 peptide, an active subfragment of tumstatin, exerts cytoprotective effects on H2O2-induced apoptosis through the inhibition of intracellular reactive oxygen species (ROS) production in H9c2 cardiomyoblasts. In this study, we investigated whether T3 peptide has cardioprotective effects against I/R injury by using in vitro and ex vivo experimental models. H9c2 cardiomyoblasts were stimulated with oxygen and glucose deprivation (OGD) for 12 h followed by reoxygenation for 1-8 h (OGD/R; in vitro model). The cells were treated with T3 peptide (30-1000 ng/ml) during OGD. Ten minutes after the pre-perfusion of T3 peptide (300 ng/ml), Langendorff perfused rat hearts were exposed to ischemia for 30 min followed by reperfusion for 1 h (ex vivo model). T3 peptide inhibited OGD/R-induced apoptosis through the inhibition of mitochondrial ROS production and dysfunction in H9c2 cardiomyoblasts. T3 peptide also prevented I/R-induced cardiac dysfunction, arrhythmia and myocardial infarction in the perfused rat heart. In conclusion, we for the first time demonstrated that T3 peptide exerts cardioprotective effects against I/R injury.
    Keywords:  Ischemia/reperfusion injury; Langendorff heart; Reactive oxygen species; Tumstatin
    DOI:  https://doi.org/10.1016/j.jphs.2019.01.010
  30. Nutr Metab (Lond). 2019 ;16 14
      Background: Adiponectin exerts several beneficial cardiovascular effects, however their specific molecular mechanisms require additional understanding. This study investigated the mechanisms of adiponectin action in the heart during high fat diet (HFD) feeding or in palmitate (PA) treated H9c2 cardiomyoblasts.Methods: 6-week-old male adiponectin knock out (Ad-KO) mice were fed chow or 60% HFD for 6 weeks then received saline or recombinant adiponectin (3μg/g body weight) for an additional 2 weeks. After acute insulin stimulation (4 U/kg), tissue and serum samples were collected for analysis. H9c2 cardiomyocytes were treated ±0.1 mM PA, the adiponectin receptor agonist AdipoRon, or the antioxidant MnTBAP then assays to analyze reactive oxygen species (ROS) production and cell death were conducted. To specifically determine the mechanistic role of S1P, gain and loss of function studies were conducted with adding S1P to cells or the inhibitors THI and SKI-II, respectively.
    Results: HFD feeding induced cardiac insulin resistance in Ad-KO mice, which was reversed following replenishment of normal circulating adiponectin levels. In addition, myocardial total triglyceride was elevated by HFD and lipidomic analysis showed increased levels of ceramides and sphingosine-1-phosphate (S1P), with only the latter being corrected by adiponectin administration. Similarly, treatment of H9C2 cardiomyoblasts with PA led to a significant increase of intracellular S1P but not in conditioned media whereas AdipoRon significantly increased S1P production and secretion from cells. AdipoRon or the antioxidant MnTBAP significantly reduced PA-induced cell death. Gain and loss of function studies suggested S1P secretion and autocrine receptor activation mediated the effect of AdipoRon to attenuate PA-induced ROS production and cell death.
    Conclusion: Our data establish adiponectin signaling-mediated increase in S1P secretion as a mechanism via which HFD or PA induced cardiomyocyte lipotoxicity, leading to insulin resistance and cell death, is attenuated.
    Keywords:  Adiponectin; Cardiomyocyte apoptosis; Ceramide; High fat diet; Palmitate; ROS; Sphingosine-1-phosphate
    DOI:  https://doi.org/10.1186/s12986-019-0342-y
  31. Cancer Lett. 2019 Mar 05. pii: S0304-3835(19)30124-7. [Epub ahead of print]
      Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers because it is highly resistant to every available therapeutic strategy, in particular conventional chemotherapy or radiotherapy (RT). Sensitizing tumor cells to existing treatments remains a good option to obtain fast and applicable results. Considering that ionizing radiations induce radiolysis-derived reactive oxygen species (ROS), we hypothesized that ROS-inducing bioactive food components (BFCs) could exacerbate ROS-related cell damages, including DNA double stranded breaks (DSBs), leaving the cellular ROS scavenging systems overwhelmed, and precipitating tumor cell death. Combination of resveratrol and capsaicin radiosensitized radiosensitive tumor cells, but RT did not increase BFC combination toxicity in radioresistant tumor cells. BFC addition to RT increased ROS production and led to significant tumor volume reduction in xenografted mouse preclinical model. Strikingly, BFCs inhibited RT-induced DNA damage molecular response by strongly limiting the first steps of DSB repair, and by keeping cells in cell cycle, provoking exacerbated intrinsic apoptosis. This study positions BFCs as potent radiosensitizers when combined, and identifies an actionable molecular pathway by resveratrol and capsaicin combination.
    Keywords:  Capsaicin; DNA repair; Pancreatic Ductal Adenocarcinoma; Radiotherapy; Resveratrol
    DOI:  https://doi.org/10.1016/j.canlet.2019.02.038
  32. Int J Biol Macromol. 2019 Mar 04. pii: S0141-8130(19)30247-8. [Epub ahead of print]
      The aim of the present investigation is to explore the innovative platform for the synthesis of plant-based nanoparticles, which contain biocompatible and biodegradable carrier of chitosan loaded with phloretin hydrophobic phytochemical applied as a stable anticancer agent. Treatment of cancer uses chemotherapeutic drugs as the cells are resistant to other drugs. However, the usage of therapeutic drug is limited by its poor solubility and low bioavailability. To overcome this problem, we fabricated the phloretin loaded chitosan nanoparticles (PhCsNPs) and physicochemical properties of PhCsNPs were characterized by FTIR, XRD, DLS, SEM and TEM. The findings indicated that the synthesized PhCsNPs were spherical and homogeneous in shape with the size distribution of 80-100 nm and exhibited stability in ultimate drug releasing profile. Further, we substantiated the anticancer efficiency of PhCsNPs through bio-assessment, such as cytotoxicity measurement, intracellular ROS, mitochondrial dysfunction, lipid peroxidation measurement, antioxidants status, apoptotic associated gene expression profile and cell cycle analysis in human oral cancer cell lines. The findings suggested that PhCsNPs augmented the mitochondrial-mediated apoptotic mechanism through the stimulation of oxidative stress, depletion of cellular antioxidants and cell cycle arrest. Our data suggested that PhCsNPs could be used as an efficient therapeutic agent for the treatment of oral cancer.
    Keywords:  Apoptosis; Oxidative stress; Phloretin
    DOI:  https://doi.org/10.1016/j.ijbiomac.2019.03.031
  33. Biomed Pharmacother. 2019 Mar;pii: S0753-3322(18)38065-X. [Epub ahead of print]111 901-908
      AIMS: Bisphenol A (BPA) can induce intestinal epithelial cell barrier dysfunction; however, its effects on the intestinal mucus barrier remain unclear. We used LS174T cells as a model to investigate the effects of BPA on the functions of intestinal goblet cells.MAIN METHODS: This study used CCK-8, flow cytometry, ELISA and real-time PCR to investigate the effects of BPA on mitochondrial dynamics, oxidative stress and apoptosis in goblet cells. In addition, mucin synthesis and secretion were evaluated using PAS staining and a PAS assay, respectively.
    KEY FINDINGS: Our results indicate that BPA reduced cell viability in a time- and concentration-dependent manner. BPA induced mitochondrial dysfunction, as indicated by the depolarization of the mitochondrial membrane potential, inhibition of mitochondrial respiratory chain complex enzyme activity and reduction of ATP production. Moreover, BPA caused oxidative stress by significantly increasing the accumulation of ROS, as well as oxidative stress products, and reducing the antioxidant capacity. Furthermore, BPA induced intestinal goblet cell apoptosis, accompanied by increased DNA fragmentation, caspase-3, -8, -9,-10 gene expression and enzyme activity. Additionally, BPA inhibited the synthesis and secretion of mucin 2.
    SIGNIFICANCE: Our data suggest that BPA affected the secretory function of intestinal goblet cells by inducing mitochondrial dysfunction, oxidative stress, and apoptosis.
    Keywords:  Bisphenol A; Intestinal goblet cells; Mitochondria; Mucin; Oxidative stress
    DOI:  https://doi.org/10.1016/j.biopha.2019.01.007
  34. J Appl Toxicol. 2019 Mar 03.
      Toxic trace metals are widespread contaminants that are potentially immunotoxic even at environmentally low exposure levels. They can modulate the immunity to infections, e.g., in wildlife species living in contaminated areas. The diverse immune cell types can be differentially affected by the exposure leading to the modulation of specific protective mechanisms. Macrophages and mast cells, part of the innate immune system, trigger immune responses and perform particular effector functions. The present study compared toxicological and functional effects of cadmium in two models of murine macrophages (RAW264.7 and NR8383 cell lines) and two models of murine mast cells (MC/9 and RBL-2H3 cell lines). Cadmium was selected as a model compound because its known potential to induce reactive oxygen species and its relevance as an environmental contaminant. Mechanisms of toxicity, such as redox imbalance and apoptosis induction were measured in stationary cells, while functional outcome effects were measured in activated cells. Cadmium-depleted glutathione antioxidant in all four cell lines tested although reactive oxygen species was not significantly increased. Mast cells had full dose-response depletion of glutathione below cytotoxic levels while in macrophages the depletion was not complete. Functional endpoints tumour necrosis factor-alpha and nitrite production in lipopolysaccharide-activated macrophages were increased by cadmium exposure. In contrast, mast cell lipopolysaccharide-induced tumour necrosis factor-alpha and IgE-mediated histamine release were reduced by cadmium. These data indicate potentially differential effects of cadmium among murine innate immune cell types, where mast cells would be more susceptible to oxidative stress and their function might be at a higher risk to be modulated compared to macrophages.
    Keywords:  TNFα; cadmium; glutathione (GSH); histamine; innate immunity; nitrite; oxidative stress
    DOI:  https://doi.org/10.1002/jat.3788
  35. Neurosci Lett. 2019 Feb 28. pii: S0304-3940(19)30130-2. [Epub ahead of print]
      Parkinson's disease (PD) is a most common progressive neurodegenerative disease mainly occurring in the elderly. Plenty of miRNAs are reported to involve in the progression of PD. However, the role of miR-9-5p in the regulation of PD pathogenesis remains unclear. The expressions of miR-9-5p and Sirtuin 1 (SIRT1) at mRNA and protein levels were determined by qRT-PCR and western blotting (WB) analyses. Cell viability and apoptosis were evaluated by MTT and flow cytometry. The levels of apoptosis-related proteins Bcl-2, Bax, Caspase-3 were detected by WB analysis. The releases of inflammatory cytokines IL-1β and TNF-α were examined by ELISA assay. ROS generation, LDH and SOD activity were evaluated using commercially available kits. Bioinformatics analysis, luciferase reporter, and qRT-PCR assays were performed to demonstrate the true interaction between miR-9-5p and SIRT1. Results showed miR-9-5p was upregulated and SIRT1 was downregulated in MPP+-treated SH-SY5Y cells in dose- and time- dependent manners. miR-9-5p knockdown attenuated MPP+-induced neurotoxicity in SH-SY5Y cells, as evidenced by the enhancement in cell viability, and the suppression in cell apoptosis, inflammation, and oxidative stress. SIRT1 was identified to be a target of miR-9-5p. Restoration of miR-9-5p aggravated SIRT1-attenuated neurotoxicity in MPP+-treated SH-SY5Y cells. Our data suggested these data indicated that miR-9-5p exerted a neurotoxic role in MPP+-derived PD by directly targeting STAT1, providing a potential therapeutic strategy for patients troubled by PD.
    Keywords:  Parkinson’s disease; Sirtuin 1; miR-9-5p; neurotoxicity
    DOI:  https://doi.org/10.1016/j.neulet.2019.02.038
  36. Toxicology. 2019 Mar 04. pii: S0300-483X(18)30393-7. [Epub ahead of print]
      Manganese (Mn) is an essential nutrient element. However, Mn is causing great environmental and occupational exposure health risk concern globally, even high rate of alcohol consumption. There is dearth of scientific information on the interaction of manganese (Mn) and ethanol (EtOH) on hippocampal functions. This study was designed to investigate the effect of EtOH on Mn - induced hippocampal toxicity with special reference to spatial learning and memory and its underlying mechanism in adults male Wistar Rats. Rats were exposed to Mn alone at 30 mg/kg or co-expose with EtOH at 1.25 and 5 g/kg body weight by oral gavage for 35 consecutive days. Morris Water Maze task was used to assessed spatial learning and memory. Subsequently, oxidative/nitrosative stress, neuro-inflammation (myeloperoxidase and cyclooxygenase-2) and protein expression of apoptotic proteins (p53 and Bax), active executioner caspase (caspase-3) and B - cell lymphoma - 2 (Bcl - 2) markers in the hippocampus were investigated. The results indicate that Mn and EtOH exposure induces spatial learning and memory deficits, increase oxidative/nitrosative stress, neuro-inflammation resulting in enhanced hippocampal apoptosis. Moreover, the results indicated that Mn co-exposure with EtOH at 1.25 and 5 g/kg body weight further exacerbates neurotoxicity in rat hippocampus when compared with single dose of Mn and EtOH alone. Collectively, EtOH increases Mn - induced oxidative/nitrosative stress, neuro-inflammation and hippocampal apoptosis via mechanism involving oxidative damages of cellular constituents, neuronal inflammation and subsequent upregulation of Bax and caspase-3 and downregulation of Bcl-2 protein expression via p53 dependent/independent pathways to induced hippocampal apoptosis associated with impaired spatial learning and memory.
    Keywords:  Manganese; Morris; ethanol; hippocampal apoptosis; neuro-inflammation; oxidative/nitrosative stress; water maze
    DOI:  https://doi.org/10.1016/j.tox.2019.03.001
  37. Cell Biol Int. 2019 Mar 06.
      Numerous studies have demonstrated the therapeutic effect of bone mesenchymal stem cells on Spinal cord injury (SCI), especially on neural stem cells (NSCs). However, the predominant mechanisms of bone mesenchymal stem cell (BMSCs) are unclear. Recently, some researchers have found that paracrine signaling plays a key role in the therapeutic capacity of BMSCs and emphasized that the protective effect of BMSCs may be due to paracrine factors. In this study, we aimed to investigate the potential mechanisms of BMSCs recover function to NSCs.NSCs were identified by immunocytochemistry. The oxidative stress environment was simulated by H2O2(50µM, 100µM, 200µM) for 2h. The apoptotic rate of the NSCs was detected via flow cytometry. Lactate dehydrogenase (LDH), malondialdehyde (MDA) and superoxide dismutase (SOD) activity were evaluated via corresponding assay kits. Western blot were used to detect the expressions of Notch1, HES1, Caspase-3, cleave Caspase-3, Bax and Bcl-2. We found that H2O2 could significantly induce the apoptosis of NSCs, increase LDH, MDA levels and decrease SOD activity by activating the Notch1 signaling pathway. DAPT (the specific blocker of Notch1) and BMSC-CM could significantly prevent the apoptotic effect and oxidative stress injury on NSCs that were treated with H2O2. We also revealed that BMSC-CM could decrease the expression of Notch1, Hes1, cleave Caspase-3, Bax and increases the expression of Bcl-2 in NSCs which was induced by H2O2. These results have revealed that BMSC-CM can neutralize the effect against oxidative stress injury on the apoptosis of NSCs by inhibiting the Notch1 signaling pathway.
    Keywords:  BMSC-CM; H2O2; NSCs; Notch1; apoptosis
    DOI:  https://doi.org/10.1002/cbin.11126
  38. Neurotox Res. 2019 Mar 04.
      Neurodegeneration in diabetic retina has been widely considered as initiating factor that may lead to vascular damage, the classical hallmark of diabetic retinopathy. Diabetes induced altered glutamate metabolism in the retina, especially through glutamate excitotoxicity might play a major role in the neurodegeneration. Increased level of branched chain amino acids (BCAAs) measured in diabetic retina might cause an increase in the neurotoxic level of glutamate by transamination of citric acid cycle intermediates. In order to analyze the transamination of BCAAs and their influence on neurodegenerative factors, we treated streptozotocin-induced diabetic rats with gabapentin, a leucine analogue and an inhibitor of branched chain amino transferase (BCATc). Interestingly, gabapentin lowered the retinal level of BCAAs in diabetic rats. Furthermore, gabapentin treatments ameliorated the reduced antioxidant glutathione level and increased malondialdehyde (MDA), the marker of lipid peroxidation in diabetic rat retinas. In addition, gabapentin also reduced the expression of proapoptotic caspase-3, a marker of apoptosis and increased anti-apoptotic marker Bcl-2 in diabetic retinas. Thus, these results suggest that gabapentin stimulates glutamate disposal, and ameliorates apoptosis and oxidative stress in diabetic rat retina. The influence of gabapentin may be due to its capacity to increase the ratio of BCKA to BCAA which in turn would reduce glutamate excitotoxicity in diabetic retina.
    Keywords:  Diabetes; Gabapentin; Glutamate; Neurodegeneration; Retina
    DOI:  https://doi.org/10.1007/s12640-019-00018-w
  39. Biosci Biotechnol Biochem. 2019 Mar 05. 1-10
      Our previous work has reported an anti-proliferative compound from moutan cortex, paeoniflorigenone which can induce cancer-selective apoptosis. However, its anti-proliferative mechanism is still unknown. According to morphology changes (hypertrophy and flattening), we hypothesized that PFG can induce senescence or inhibit cell mitosis. Here we show that PFG can induce cellular senescence, evidenced by the expression of senescence-associated β-galactosidase, G0/G1 cell cycle arrest and permanent loss of proliferative ability, in normal TIG-1 diploid fibroblast but not cancerous HeLa cells. In cancerous HeLa cells, PFG inhibited proliferation by inducing S and G2/M cell cycle arrest and mitosis inhibition. DNA damage response was activated by PFG, interestingly the reactive oxygen species level was suppressed instead of escalated. To sum up, we report 3 new roles of PFG as, 1. inducer of premature senescence in normal TIG-1 cells, 2. inhibitor of mitosis in cancerous HeLa cells, 3. ROS scavenger. Abbreviations: PFG: Paeoniflorigenone; ROS: reactive oxygen species; ATM: ataxia telangiectasia mutated; t-BHP: tert-butyl hydroperoxide; SA-β-gal: senescence-associatedβ-galactosidase; DNA-PKcs: DNA-dependent protein kinase; γ-H2AX: H2AX phosphoryla-tion at Ser-139.
    Keywords:  Paeoniflorigenone; anti-proliferative; cell cycle arrest; reactive oxygen species; senescence
    DOI:  https://doi.org/10.1080/09168451.2019.1585744
  40. Food Chem Toxicol. 2019 Feb 28. pii: S0278-6915(19)30092-4. [Epub ahead of print]126 277-284
      Alcoholic liver disease (ALD), as one of the most common diseases, has become a global threat to human health. The aim of this study was designed to investigate the hepatoprotective effects of ginsenoside Rk3 against ALD and to discover the potential mechanisms of these protective effects. Mice were intragastrically administered 50% alcohol and treated with ginsenoside Rk3 (25 and 50 mg/kg) once per day for 6 weeks. The results indicated that ginsenoside Rk3 promoted hepatic function through significant downgrading AST and ALT levels in the serum, attenuating oxidative stress, and restoring antioxidant balance in hepatic tissue. Additionally, ginsenoside Rk3 significantly reduced the expression of inflammatory cytokines, such as NF-κB, TNF-α, IL-6, and IL-1β in the mice. Furthermore, ginsenoside Rk3 supplementation significantly inhibited apoptotic protein expression in the liver. The present study clearly demonstrates that ginsenoside Rk3 exerts a protective effect against ALD-induced liver injury because of its antioxidant, anti-apoptotic, and anti-inflammatory activities. The findings from the present investigation show that ginsenoside Rk3 might be a promising candidate treatment agent against ALD.
    Keywords:  Alcoholic liver disease; Apoptosis; Ginsenoside Rk3; Inflammation; Oxidation
    DOI:  https://doi.org/10.1016/j.fct.2019.02.032
  41. J Med Chem. 2019 Mar 07.
      Poly(ADP-ribose) polymerase-1 (PARP-1) is a new potential target for anticancer-drug discovery. A series of bromophenol-thiosemicarbazone hybrids as PARP-1 inhibitors were designed, synthesized and evaluated for their antitumor activities. Among them, the most promising compound, 11, showed excellent selective PARP-1 inhibitory activity (IC50 = 29.5 nM) over PARP-2 (IC50 > 1000 nM) and potent anticancer activities toward the SK-OV-3, Bel-7402 and HepG2 cancer cell lines (IC50 = 2.39, 5.45 and 4.60 µM), along with inhibition of tumor growth in an in vivo SK-OV-3 cell xenograft model. Further study demonstrated that compound 11 played an anti-tumor role through multiple anticancer mechanisms, including the induction of apoptosis and cell-cycle arrest, cellular accumulation of DNA double-strand breaks, DNA repair alterations, inhibition of H2O2-triggered PARylation, antiproliferative effects via the production of cytotoxic reactive oxygen species (ROS) and autophagy. In addition, compound 11 displayed good pharmacokinetic characteristics and favorable safety. These observations demonstrate that compound 11 may serve as a lead compound for the discovery of new anticancer drugs.
    DOI:  https://doi.org/10.1021/acs.jmedchem.8b01946
  42. Biol Trace Elem Res. 2019 Mar 08.
      Plant-derived synthesis of silver nanoparticles (AgNPs) has found wide biomedical applications including cancer cure. This report deals with biosynthesis of silver nanoparticles (MZLAgNPs) employing leaf extracts of Manilkara zapota (L.) under optimized conditions. Characterization of MZLAgNPs using UV-Vis spectroscopy, FTIR, XRD, and FESEM analyses revealed that the particles were predominantly spherical averaging 24 nm in size. Their cellular effects were assessed by MTT assay, fluorescence, and scanning electron microscopy of cells stained with propidium iodide, acridine orange/ethidium bromide, and annexin V-FITC to visualize signs of apoptosis. Evaluation of cell proliferation by clonogenic assay, wound healing ability by scratch assay and cell cycle distribution by flow-cytometry was also carried out. Apoptosis-related gene expressions were analyzed by RTq-PCR and western blot analysis. MZLAgNPs selectively inhibited growth of colorectal carcinoma HCT116, HeLa, and non-small lung carcinoma A549 cells, dose-dependently with IC50 concentrations of 8, 16, and 29 μg/mL respectively, following 72-h treatment, without affecting growth of normal human lymphocytes and erythrocytes. Apoptosis induction was observed by fluorescence and scanning electron microscopy. Overproduction of reactive oxygen species (ROS), reduction of mitochondrial membrane potential, upregulation of apoptotic-related genes - PUMA, cas-3, cas-8, cas-9, and BAX, expression of caspase 3, and occurrence of PARP cleavage were observed in MZLAgNPs/cisplatin treated cells. Taken together, our results clearly demonstrate the therapeutic potential of biogenic MZLAgNPs as an effective agent for killing colorectal carcinoma cells by apoptosis induction.
    Keywords:  Apoptosis; Colorectal carcinoma; Manilkara zapota; Silver nanoparticles
    DOI:  https://doi.org/10.1007/s12011-019-1653-6
  43. Cells. 2019 Mar 05. pii: E218. [Epub ahead of print]8(3):
      A compound isolated from Glycyrrhiza uralensis, licochalcone A (LA) exhibits anti-inflammatory and anti-tumor properties in various cell lines. LA has been found to promote autophagy and suppress specificity protein 1, inducing apoptosis in breast cancer cells. However, the regulation of breast cancer cell invasion and migration by LA is elusive. Thus, the present study investigated whether LA induces apoptosis and cellular motility in MDA-MB-231 breast cells, and investigated the underlying molecular mechanisms. MDA-MB-231 cells treated with LA and cell viability measured by cell counting kit-8 assay. Apoptotic signal proteins checked by flow cytometry, fluorescent staining, and Western blot. LA effectively suppressed cell migration, and modulated E-cadherin and vimentin expression by blocking MAPK and AKT signaling. LA inhibited cell proliferation and cell cycle, modulated mitochondrial membrane potential and DNA damage, and reduced oxidative stress in MDA-MB-231 cells. LA also activated cleaved-caspase 3 and 9, significantly decreased Bcl-2 expression, ultimately causing the release of cytochrome c from the mitochondria into the cytoplasm. Overall, our findings suggest that LA decreases cell proliferation and increases reactive oxygen species production for induced apoptosis, and regulates E-cadherin and vimentin by reducing MAPK and AKT signaling, resulting in suppressed MDA-MB-231 cell migration and invasion.
    Keywords:  MDA-MB-231 cells; apoptosis; caspase-3; cellular motility; licochalcone A
    DOI:  https://doi.org/10.3390/cells8030218
  44. Front Mol Neurosci. 2019 ;12 7
      The noradrenaline (NA) level in the brain is reduced during rapid eye movement sleep (REMS). However, upon REMS deprivation (REMSD) its level is elevated, which induces apoptosis and the degeneration of neurons in the brain. In contrast, isolated studies have reported that NA possesses an anti-oxidant property, while REMSD reduces lipid peroxidation (LP) and reactive oxygen species (ROS). We argued that an optimum level of NA is likely to play a physiologically beneficial role. To resolve the contradiction and for a better understanding of the role of NA in the brain, we estimated LP and ROS levels in synaptosomes prepared from the brains of control and REMS deprived rats with or without in vivo treatment with either α1-adrenoceptor (AR) antagonist, prazosin (PRZ) or α2-AR agonist, clonidine (CLN). REMSD significantly reduced LP and ROS in synaptosomes; while the effect on LP was ameliorated by both PRZ and CLN; ROS was prevented by CLN only. Thereafter, we evaluated in vitro the effects of NA, vitamin E (Vit E), vitamin C (Vit C), and desferrioxamine (DFX, iron chelator) in modulating hydrogen peroxide (H2O2)-induced LP and ROS in rat brain synaptosomes, Neuro2a, and C6 cells. We observed that NA prevented ROS generation by chelating iron (inhibiting a Fenton reaction). Also, interestingly, a lower dose of NA protected the neurons and glia, while a higher dose damaged the neurons and glia. These in vitro and in vivo results are complementary and support our contention. Based on the findings, we propose that REMS maintains an optimum level of NA in the brain (an antioxidant compromised organ) to protect the latter from continuous oxidative onslaught.
    Keywords:  C6; Neuro2a; catecholamine; cell death; lipid peroxidation; oxidative stress; synaptosome; vitamins C and E
    DOI:  https://doi.org/10.3389/fnmol.2019.00007
  45. Front Pharmacol. 2019 ;10 101
      Background: Fibroblast growth factor 21 (FGF21), a member of a family of atypical FGFs, functions as cytokine to control endocrinology and metabolism. Recently, the roles of FGF21 in cardio-cerebral-vascular diseases have been gradually uncovered. In the present study, we investigated the effect of FGF21 on bEnd.3 cerebral microvascular endothelial cells (CMECs) upon hypoxia stress. Methods and Results: CMECs were cultured in the condition of 1% O2 for 8 h to induce hypoxia stimuli. For FGF21 treatment, recombinant FGF21 (50 nM) was added into the culture medium. Various biomedical assays were used to evaluate the hypoxia-induced injury in CMECs. Under normoxia condition, FGF21 had no obvious effect on cell viability and did not cause any cytotoxicity on CMECs. Under hypoxia condition, FGF21 significantly attenuated the hypoxia-induced injury, evidenced by the influences of FGF21 on CMEC viability and LDH release. TUNEL staining assay and immunoblotting of caspase-3 showed that FGF21 reduced hypoxia-induced apoptosis in CMECs. Mechanistically, FGF21 treatment compromised the hypoxia-induced changes of reactive oxygen species, malondialdehyde, total antioxidant activity, and total superoxide dismutase levels. FGF21 administration decreased hypoxia-induced matrix metalloprotein 3 and matrix metalloprotein 2/9 activity in CMECs. Activities of cyclooxygenase-2 and NF-κB-p65, two pro-inflammatory factors, were also upregulated by hypoxia but suppressed by FGF21. At last, we found that FGF21 increased heat shock protein family A member 1A (HSP72) mRNA and protein expression. Blockade of HSP72 by a pharmacological inhibitor VER155008 or specific siRNA-mediated knockdown abrogated the protection of FGF21 against hypoxia in CMECs. Conclusion: These data demonstrate that FGF21 protects against hypoxia stress-induced injury in CMECs by inducing HSP72 expression, suggesting a therapeutic value of FGF21 in hypoxia-related brain diseases such as ischemic stroke and acute mountain sickness.
    Keywords:  FGF21; NF-κB; cerebral microvascular endothelial cell; cyclooxygenase-2; heat shock protein family A member 1A; hypoxia; matrix metalloprotein; oxidative
    DOI:  https://doi.org/10.3389/fphar.2019.00101
  46. J Bioenerg Biomembr. 2019 Mar 07.
      Despite radiotherapy is an effective regimen in cancer treatment, resistance to tumor therapy still is a major challenge to radiotherapy and results in cancer recurrence and metastasis. Then the sensitization of tumor cells to ionizing radiation (IR) would be beneficial in cancer treatment. The aim of this study was to evaluate the synergistic effect of mefenamic acid (MEF) on colon cancer cell (HT-29) exposure to IR. HT-29 cells were treated with MEF and then exposed to IR. The synergistic effect of MEF is evaluated by clonogenic assay and flow cytometry. The productions of reactive oxygen species (ROS) were determined in irradiated and treated cells with MEF. The findings of this study showed that MEF had anti-cancer effect on colon cancer cell line and it increased the apoptosis in irradiated HT-29 cells. Also MEF reduced the number of cell colonies when HT-29 cells pre-treated with MEF and irradiated. MEF increased ROS production in irradiated cells. This additive effect of MEF with IR in killing of HT-29 cell was observed at low (10 μM) and medium (100 μM) concentrations of MEF. The present study demonstrates that MEF to be an additive effect on apoptosis and cell death induced by IR in colon cancer cells.
    Keywords:  Apoptosis; Ionizing radiation; Mefenamic acid; Synergistic, radiosensitizing
    DOI:  https://doi.org/10.1007/s10863-019-09792-w
  47. Eur J Heart Fail. 2019 Mar 06.
      AIMS: Butyric acid (BUT), a short chain fatty acid produced daily by the gut microbiota, has proven beneficial in models of cardiovascular diseases. With advancements in cancer survival, an increasing number of patients are at risk of anticancer drug cardiotoxicity. Here we assess whether the novel BUT derivative phenylalanine-butyramide (FBA) protects from doxorubicin (DOXO) cardiotoxicity, by decreasing oxidative stress and improving mitochondrial function.METHODS AND RESULTS: In C57BL6 mice, DOXO produced left ventricular dilatation assessed by echocardiography. FBA prevented left ventricular dilatation, fibrosis and cardiomyocyte apoptosis when co-administered with DOXO. DOXO increased atrial natriuretic peptide, brain natriuretic peptide, connective tissue growth factor, and matrix metalloproteinase-2 mRNAs, which were not elevated on co-treatment with FBA. DOXO, but not FBA + DOXO mice, also showed higher nitrotyrosine levels, and increased inducible nitric oxide synthase expression. Accordingly, DOXO hearts showed lower levels of intracellular catalase vs. sham, while pre-treatment with FBA prevented this decrease. We then assessed for reactive oxygen species (ROS) emission: DOXO induced increased activity of mitochondrial superoxide dismutase and higher production of H2 O2 , which were blunted by FBA pre-treatment. FBA also ameliorated mitochondrial state 3 and state 4 respiration rates that were compromised by DOXO. Furthermore, in DOXO animals, the mitochondrial degree of coupling was significantly increased vs. sham, while FBA was able to prevent such increase, contributing to limit ROS production, Finally, FBA reduced DOXO damage in human cellular models, and increased the tumour-killing action of DOXO.
    CONCLUSIONS: Phenylalanine-butyramide protects against experimental doxorubicin cardiotoxicity. Such protection is accompanied by reduction in oxidative stress and amelioration of mitochondrial function.
    Keywords:  Doxorubicin cardiotoxicity; Heart failure; Oxidative and nitrosative stress; Phenylalanine-butyramide
    DOI:  https://doi.org/10.1002/ejhf.1439
  48. Toxicol Appl Pharmacol. 2019 Mar 01. pii: S0041-008X(19)30073-0. [Epub ahead of print]
      BACKGROUND AND PURPOSE: Doxorubicin (Dox)-induced cardiotoxicity limits its clinical use. A number of microRNAs (miRs) have been found essential in Dox-induced cardiotoxicity. The aim of the present study was to elucidate the effects of miR-23a on Dox-induced cardiomyocyte apoptosis and underlying mechanisms.EXPERIMENTAL APPROACH: Dox-induced cardiotoxicity model was established in primary neonatal rat ventricular myocytes (NRVMs). MTT assay, Live/Dead staining was employed to examine the viability and cell death of NRVMs. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) were measured. Protein levels of mitochondria biogenesis and fission/fusion associated factors including peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), dynamin-related protein-1 (Drp1) and mitofusin 2 (Mfn2) were detected. Meanwhile, apoptosis-related cytochrome c (Cyt c) and caspase-3 expression were examined by western blot. PGC-1α siRNA was employed to validate the role of miR-23a in Dox-induced cardiotoxicity.
    KEY RESULTS: MiR-23a expression was significantly increased by Dox concentration-dependently. Inhibition of miR-23a markedly increased viability and MMP, reduced cell death and ROS production of NRVMs. MiR-23a mimic significantly inhibited expression of its target PGC-1α. MiR-23a inhibitor significantly diminished phosphorylation of Drp1 without affecting Mfn2 expression. Protein expression of Cyt c and cleaved caspase-3 were markedly inhibited by miR-23a inhibitor. The protective effects of miR-23a inhibitor were reversed by PGC-1α siRNA.
    CONCLUSIONS AND IMPLICATIONS: Increased miR-23a promoted mitochondrial injury in the Dox-induced cellular model. Inhibition of miR-23a attenuated cardiomyocyte damage by directly targeting PGC-1α/p-Drp1, thereby inhibiting mitochondria-dependent apoptosis. These findings may provide a new potential target for the treatment of Dox-induced cardiotoxicity.
    Keywords:  Doxorubicin; Drp1; Mitochondria; PGC-1α; miR-23a
    DOI:  https://doi.org/10.1016/j.taap.2019.02.016
  49. Nanotechnology. 2019 Mar 08.
      Circulating tumor cells (CTCs) were kind of rare cells that firstly shed from solid tumors and then existed in the bloodstream. The effective capture and separation of CTCs have significant meanings in cancer's diagnosis and prognosis. In this study, novel Fe3O4-FePt magnetic nanocomposites (Fe3O4-FePt MNCs) were constructed by integrating face centered cubic (fcc) FePt nanoparticles (NPs) onto the surface of the Fe3O4@SiO2 core. After further modified with NH2-PEG-COOH and tumor-targeting molecule tLyP-1, the acquired Fe3O4-FePt MNCs with excellent biocompatibility and stability could efficiently target and capture tLyP-1 receptor-positive CTCs. Based on the acidic microenvironment within cancer cells, the FePt layer could rapidly release active Fe2+ ions, which could catalyze H2O2 into reactive oxygen species (ROS) and further induce cancer cells in-situ apoptosis while no distinct cytotoxicity to normal cells. Moreover, the Fe3O4@SiO2 core with intrinsic magnetism had huge potential in bioseparation of CTCs. The in vitro ROS fluorescence imaging experiments and cell capture and separation experiments indicated that the Fe3O4-FePt MNCs could specifically capture and separated cancer cells in CTCs model and further induce in-situ apoptosis. Therefore, the Fe3O4-FePt MNCs could serve as a promising multifunctional nanoseparator for efficiently capturing CTCs and simultaneously inducing in-situ chemotherapy.
    Keywords:  Capture and Separation; Circulating Tumor Cells; In-situ Chemotherapy; Magnetic Nanocomposites
    DOI:  https://doi.org/10.1088/1361-6528/ab0e25
  50. Chem Biol Interact. 2019 Mar 02. pii: S0009-2797(18)31628-4. [Epub ahead of print]
      The cannabinoid type 2 receptor (CB2) has recently emerged as an important therapeutic target for cancer as well as cardiovascular diseases. The CB2 receptor downregulation has been reported in solid tumors and cardiovascular diseases, therefore the CB2 receptor activation has been considered as a viable strategy for chemotherapy as well as cardioprotection. In chemotherapy, doxorubicin (DOX) is an important drug that continues to be the mainstay of chemotherapy in solid tumors, leukemia, and lymphoma. However, the use of DOX is often limited due to its lethal cardiotoxicity. Considering the role of CB2 receptors in cardiovascular diseases and cancer, the activation of CB2 receptors may protect against DOX-induced chronic cardiotoxicity in rats. In the present study, we investigated the cardioprotective effect of a selective CB2 receptor agonist; β-Caryophyllene (BCP), a natural bicyclic sesquiterpene, against DOX-induced chronic cardiotoxicity in rats. AM630, a CB2 receptor antagonist was administered as a pharmacological challenge prior to BCP treatment to demonstrate CB2 receptor mediated cardioprotective mechanism of BCP. DOX (2.5 mg/kg) was injected intraperitoneally once a week for five weeks to induce chronic cardiotoxicity in rats. BCP was also injected into rats six days a week for a total duration of five weeks. DOX induced a significant decline in cardiac function and oxidative stress evidenced by the depletion of antioxidant enzymes, glutathione, and increased lipid peroxidation. DOX also triggered activation of nuclear factor kappa B (NF-κB) and increased the levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and expression of the inflammatory mediators (iNOS and COX-2) in the heart. Furthermore, DOX also upregulated the expression of pro-apoptotic markers such as Bax, p53, cleaved PARP, active caspase-3 and downregulated anti-apoptotic marker Bcl-2 in the myocardium. BCP treatment exerted significant cardioprotective effect by salvaging the heart tissues, improving cardiac function, mitigating oxidative stress, inflammation, and apoptosis. The histological and ultrastructural studies also appear in line with our findings of biochemical and molecular parameters. The CB2 receptor-mediated cardioprotective mechanism was further confirmed by the abrogation of the beneficial effects of BCP with prior administration of the CB2 receptor antagonist; AM630. Our study revealed the novel mechanism of BCP in cardioprotection against DOX-induced chronic cardiotoxicity by the activation of CB2 receptors.
    Keywords:  AM630; Cannabinoid 2 receptor; Cardiotoxicity; Doxorubicin; β-Caryophyllene
    DOI:  https://doi.org/10.1016/j.cbi.2019.02.028
  51. Food Chem Toxicol. 2019 Feb 27. pii: S0278-6915(19)30091-2. [Epub ahead of print]126 262-276
      Zearalenone (ZEA), a non-steroidal estrogen mycotoxin produced by several species of Fusarium fungi, can be metabolized into many other derivatives by microorganisms, plants, animals and humans. It can affect mammalian reproductive capability by impacting the synthesis and secretion of sex hormones, including testosterone, estradiol and progesterone. This review summarizes the mechanisms in which ZEA and its derivatives disturb the synthesis and secretion of sex steroid hormones. Because of its structural analogy to estrogen, ZEA and its derivatives can exert a variety of estrogen-like effects and engage in estrogen negative feedback regulation, which can result in mediating the production of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in the pituitary gland. ZEA and its derivatives can ultimately reduce the number of Leydig cells and granulosa cells by inducing oxidative stress, endoplasmic reticulum (ER) stress, cell cycle arrest, cell apoptosis, and cell regeneration delay. Additionally, they can disrupt the mitochondrial structure and influence mitochondrial functions through overproduction of reactive oxygen species (ROS) and aberrant autophagy signaling ways. Finally, ZEA and its derivatives can disturb the expressions and activities of the related steroidogenic enzymes through cross talking between membrane and nuclear estrogen receptors.
    Keywords:  Granulosa cell; Leydig cell; Steroid sex hormones; Steroidogenesis; Zearalenone
    DOI:  https://doi.org/10.1016/j.fct.2019.02.031
  52. Environ Sci Pollut Res Int. 2019 Mar 05.
      Oxidative stress plays a significant role in the pathophysiology of numerous kidney diseases, generally mediated by reactive oxygen species (ROS). Arsenic (Ar) is known to exert its toxicity through the generation of ROS and inflammation. The current study investigates the protective effects of sulforaphane (SFN) against arsenic-induced renal damage via PI3K/Akt-mediated Nrf2 pathway signaling. Thirty-two male albino Wistar rats were randomly divided into four groups of eight animals each, designated as control, arsenic (Ar), sulforaphane plus Ar (SFN+Ar), and sulforaphane alone (SFN), with oral administration of Ar (5 mg/kg BW) and SFN (80 mg/kg BW) daily for 28 days. Ar administration significantly (P < 0.05) increased the levels of ROS, OHdG, Ar accumulation, and lipid peroxidation, and decreased levels of enzymatic and nonenzymatic antioxidants. Notably, a significant (P < 0.05) increase was observed in markers of apoptosis, DNA damage, TUNEL-positive cells, and dark staining of ICAM-1 in renal tissue with decreased PI3K/Akt/Nrf2 gene expression. The biochemical findings were supported by histopathological and electron microscopy evaluation, which showed severe renal damage in rats treated with Ar. Pretreatment with SFN significantly (P < 0.05) attenuated renal ROS, OHdG, lipid peroxidation, and DNA damage, and increased phase II antioxidants via PI3K/Akt-mediated Nrf2 activation in renal tissue. These results show that dietary supplementation with SFN protects against Ar-induced nephrotoxicity via the PI3K/Akt-mediated Nrf2 signaling pathway in the rat kidney.
    Keywords:  Arsenic; Nephrotoxicity; Nrf2 pathway; PI3K/Akt; Rat; Sulforaphane
    DOI:  https://doi.org/10.1007/s11356-019-04502-w
  53. Life Sci. 2019 Mar 05. pii: S0024-3205(19)30151-1. [Epub ahead of print]
      AIM: To investigate the cardioprotective effects of prolonged and moderate exercise training on cellular and molecular events early after myocardial infarction.MATERIALS AND METHODS: Male Wistar rats were divided in sedentary or exercised group; both groups underwent to a myocardial infarction. All the molecular and immunohistochemical analyses on hearts of sedentary and exercised rats were performed 48 h after surgical procedure. SIRT1 and SIRT3 expression were measured and two of the pathways activated by sirtuins, p53-induced apoptosis and Forkhead boxO (FOXO)3a-induced oxidative stress, were investigated. All the experiments were performed also in presence of the SIRT inhibitor, EX527.
    KEY FINDINGS: Fourty-eight hours post myocardial infarction, exercise training induced the activation of SIRT1 and SIRT3 pathway reducing cardiomyocytes apoptosis and oxidative damage. Molecular data were confirmed by immunohistochemical evaluations. These effects are more evident in border infarcted zone than in the remote myocardium.
    SIGNIFICANCE: Exercise training is a non-pharmacological prevention strategy in cardiovascular diseases and the sirtuins family seems to be as novel and attractive target in cardioprotection.
    Keywords:  Apoptosis; Exercise training; Myocardial infarction; Oxidative stress; SIRT1; SIRT3
    DOI:  https://doi.org/10.1016/j.lfs.2019.03.001
  54. Int J Biol Macromol. 2019 Mar 02. pii: S0141-8130(19)30495-7. [Epub ahead of print]
      Myocardial Infarction Associated Transcript (MIAT) is a subnuclear lncRNA that interferes with alternative splicing and is associated with increased risk of various heart conditions and nervous system tumours. The current study aims to elucidate the role of MIAT in cell survival, apoptosis and migration in neuroblastoma and glioblastoma multiforme. To this end, MIAT was silenced by MIAT-specific siRNAs in neuroblastoma and glioblastoma cell lines, and RNA sequencing together with a series of functional assays were performed. The RNA sequencing has revealed that the expression of an outstanding number of genes is altered, including genes involved in cancer-related processes, such as cell growth and survival, apoptosis, reactive oxygen species (ROS) production and migration. Furthermore, the functional studies have confirmed the RNA sequencing leads, with our key findings suggesting that MIAT knockdown eliminates long-term survival and migration and increases basal apoptosis in neuroblastoma and glioblastoma cell lines. Taken together with the recent demonstration of the involvement of MIAT in glioblastoma, our observations suggest that MIAT could possess tumour-promoting properties, thereby acting as an oncogene, and has the potential to be used as a reliable biomarker for neuroblastoma and glioblastoma and be employed for prognostic, predictive and, potentially, therapeutic purposes for these cancers.
    Keywords:  Apoptosis; Glioblastoma; MIAT; Neuroblastoma; Oxidative stress; RNA sequencing
    DOI:  https://doi.org/10.1016/j.ijbiomac.2019.03.005
  55. Phytomedicine. 2019 Jan 30. pii: S0944-7113(19)30024-8. [Epub ahead of print]58 152853
      BACKGROUND: While incidences of cancer are continuously increasing, drug resistance of malignant cells is observed towards almost all pharmaceuticals. Several isoflavonoids and flavonoids are known for their cytotoxicity towards various cancer cells.PURPOSE: The aim of this study was to determine the cytotoxicity of isoflavones: osajin (1), 5,7-dihydroxy-4'-methoxy-6,8-diprenylisoflavone (2) and biflavonoids: chamaejasmin (3), 7,7″-di-O-methylchamaejasmin (4) and campylospermone A (5), a dimeric chromene [diphysin(6)] and an ester of ferullic acid with long alkyl chain [erythrinasinate (7)] isolated from the stem bark and roots of the Kenyan medicinal plant, Ormocarpum kirkii. The mode of action of compounds 2 and 4 was further investigated.
    METHODS: The cytotoxicity of compounds was determined based on the resazurin reduction assay. Caspases activation was evaluated using the caspase-Glo assay. Flow cytometry was used to analyze the cell cycle (propodium iodide (PI) staining), apoptosis (annexin V/PI staining), mitochondrial membrane potential (MMP) (JC-1) and reactive oxygen species (ROS) (H2DCFH-DA). CCRF-CEM leukemia cells were used as model cells for mechanistic studies.
    RESULTS: Compounds 1, 2 and 4 displayed IC50 values below 20 µM towards CCRF-CEM and CEM/ADR5000 leukemia cells, and were further tested towards a panel of 7 carcinoma cells. The IC50 values of the compounds against carcinoma cells varied from 16.90 µM (in resistant U87MG.ΔEGFR glioblastoma cells) to 48.67 µM (against HepG2 hepatocarcinoma cells) for 1, from 7.85 µM (in U87MG.ΔEGFR cells) to 14.44 µM (in resistant MDA-MB231/BCRP breast adenocarcinoma cells) for 2, from 4.96 µM (towards U87MG.ΔEGFRcells) to 7.76 µM (against MDA-MB231/BCRP cells) for 4, and from 0.07 µM (against MDA-MB231 cells) to 2.15 µM (against HepG2 cells) for doxorubicin. Compounds 2 and 4 induced apoptosis in CCRF-CEM cells mediated by MMP alteration and increased ROS production.
    CONCLUSION: The present report indicates that isoflavones and biflavonoids from Ormocarpum kirkii are cytotoxic compounds with the potential of being exploited in cancer chemotherapy. Compounds 2 and 4 deserve further studies to develop new anticancer drugs to fight sensitive and resistant cancer cell lines.
    Keywords:  Apoptosis; Biflavonoid; Cancer; Isoflavone; Multi-drug resistance; Ormocarpum kirkii
    DOI:  https://doi.org/10.1016/j.phymed.2019.152853
  56. Bioorg Med Chem. 2019 Feb 20. pii: S0968-0896(18)31802-9. [Epub ahead of print]
      A series of quinizarin derivatives containing quaternary ammonium salts and/or thiourea groups were synthesized and their anticancer activities against leukemia cell lines have been tested. Results showed that most of quinizarin derivatives could inhibit the proliferation of leukemia cells. Among these derivatives, compound 3 showed good inhibition activity against various leukemia cells with IC50 values ranging from 0.90 ± 2.55 μM to 10.90 ± 3.66 μM. At the same time, compound 3 also inhibited the growth of human embryonic kidney-293 cell (HEK-293). Molt-4 and Jurkat cells, acute T lymphoblastic leukemia (T-ALL) cell lines, were selected to reveal potential anticancer mechanism of compound 3. Compound 3 inhibited the proliferation of Molt-4 and Jurkat cells in a dose- and time-dependent manner and led to a marked G0/G1 phase arrest. Analysis of Annexin V-APC and intracellular reactive oxygen species (ROS) level by flow cytometry showed that compound 3 induced significant apoptosis in Molt-4 and Jurkat cells. Western blotting assay showed that compound 3 activated the caspase-dependent apoptosis pathway and induced the degradation of Bcl-2 and c-myc protein.
    Keywords:  Apoptosis; Leukemia; Mechanism; Quinizarin derivatives; Synthesis
    DOI:  https://doi.org/10.1016/j.bmc.2019.02.041
  57. J Toxicol Sci. 2019 ;44(3): 134-144
      Tris (1,3-dichloro-2-propyl) phosphate (TDCPP) is the most widely used organophosphorus flame retardant, which is now used instead of polybrominated diphenyl ethers (PBDEs). TDCPP has frequently been detected in inorganic environmental matrices, such as soil, water and air as well as biota. In vitro effects of TDCPP on cells had not been previously elucidated. Therefore, in the present study, cytotoxicity, DNA damage, cell cycle distribution, apoptosis caused by TDCPP was studied in RAW264.7 macrophage cells. TDCPP reduced viability of RAW264.7 cells in a concentration-dependent manner and caused damage to DNA that was detected by use of the comet assay and caused up-regulation of the level of γ-H2AX. TDCPP increased the intracellular reactive oxygen species (ROS) level in RAW264.7 cells up to 1.44-fold compared to the control group at 12 hr. Percentages of cells in G1 and G2 phases of the cell cycle were dose-dependently greater in cells exposed to TDCPP. TDCPP significantly down-regulated expression of CDK-4, Cyclin D1, Cyclin B1, CDC-2, which are regulators of G1 and G2 phases of the cell cycle. These results demonstrated that TDCPP is cytotoxic and damages DNA in RAW264.7 cells, which resulted in arrest of the cell cycle at G1 and G2 phases and resulted in apoptosis, suggest the necessity to evaluate the effects of TDCPP on the immune system at the cellular level.
    Keywords:  Cell cycle arrest; DNA damage; Organophosphorus flame retardant; RAW264.7 cells; TDCPP
    DOI:  https://doi.org/10.2131/jts.44.134
  58. Biol Trace Elem Res. 2019 Mar 05.
      Iron accumulation plays a major role in neuronal cell death which has severe effects on mental health like neurodegenerative disorders. The present work aims to explore the involvement of molecular pathways involved in iron-mediated neuronal cell death using Ferric Ammonium Citrate (FAC) as a source of iron to treat neuroblastoma SH-SY5Y cells. In this study, it was found that cytotoxicity induced by iron treatment is highly correlated with enhanced intracellular reactive oxygen species (ROS) generation and loss of mitochondrial integrity. Appearance of early and late apoptotic cells with altered nuclear morphology and increased expression of effector proteins, i.e., cleaved Caspase 3 and cleaved PARP (Poly-ADP-ribose Polymerase), clearly confirmed iron-induced apoptotic cell deaths. Furthermore, excess accumulation of acidic vesicles and microtubule-associated protein 1 light chain 3 (LC3) puncta and LC3II/I expressions were observed. Simultaneously, ultrastructural studies of SH-SY5Y cells demonstrated the accumulation of a large number of autophagosomes, autophagic vacuolization, and swollen mitochondria which further confirmed the induction of autophagy concomitant with mitochondrial damage. Furthermore, increased incorporation of lysosome-specific dye, LysoTracker Deep Red, and the red fluorescence retention of LC3-GFP-RFP constructs indicates the incomplete autophagy or autophagy dysfunction due to altered lysosomal activity. Hence, the present work unveiled the interruption in autophagy progression caused by the plausible suppression of lysosomal activity due to iron treatment resulting in autophagic cell death in SH-SY5Y cell lines. In general, both apoptotic and autophagic pathways were prominent and each of the pathways played their prospective roles, in iron-mediated neuronal cell death.
    Keywords:  Apoptosis; Autophagic cell death (ACD); Autophagy; Ferric Ammonium Citrate (FAC); Lysosomal activity; SH-SY5Y
    DOI:  https://doi.org/10.1007/s12011-019-01679-6
  59. Cell Death Dis. 2019 Mar 08. 10(3): 235
      Antibody-mediated rejection (AMR) is the major cause of allograft loss after solid organ transplantation. Circulating donor-specific antibodies against human leukocyte antigen (HLA), in particular HLA class II antibodies are critical for the pathogenesis of AMR via interactions with endothelial cells (ECs). To investigate the effects of HLA class II antibody ligation to the graft endothelium, a model of HLA-DR antibody-dependent stimulation was utilized in primary human ECs. Antibody ligation of HLA class II molecules in interferon-γ-treated ECs caused necrotic cell death without complement via a pathway that was independent of apoptosis and necroptosis. HLA-DR-mediated cell death was blocked by specific neutralization of antibody ligation with recombinant HLA class II protein and by lentiviral knockdown of HLA-DR in ECs. Importantly, HLA class II-mediated cytotoxicity was also induced by relevant native allele-specific antibodies from human allosera. Necrosis of ECs in response to HLA-DR ligation was mediated via hyperactivation of lysosomes, lysosomal membrane permeabilization (LMP), and release of cathepsins. Notably, LMP was caused by reorganization of the actin cytoskeleton. This was indicated by the finding that LMP and actin stress fiber formation by HLA-DR antibodies were both downregulated by the actin polymerization inhibitor cytochalasin D and inhibition of Rho GTPases, respectively. Finally, HLA-DR-dependent actin stress fiber formation and LMP led to mitochondrial stress, which was revealed by decreased mitochondrial membrane potential and generation of reactive oxygen species in ECs. Taken together, ligation of HLA class II antibodies to ECs induces necrotic cell death independent of apoptosis and necroptosis via a LMP-mediated pathway. These findings may enable novel therapeutic approaches for the treatment of AMR in solid organ transplantation.
    DOI:  https://doi.org/10.1038/s41419-019-1319-5
  60. Iran J Basic Med Sci. 2019 Feb;22(2): 179-186
      Objectives: Cisplatin is an effective antineoplastic agent; its clinical utility, however, is limited by a few salient toxic side effects like nephrotoxicity. This study aimed to determine the potential protective effects of tangeretin, a citrus-derived flavonoid, against renal tubular cell injury in cisplatin-induced renal toxicity of rats.Materials and Methods: Tangeretin was injected intraperitoneally at 2.5 and 5 mg/kg doses for 10 days, and a single dose of cisplatin (8 mg/kg) was injected on the 7th day. Tests of kidney function and tubular injury in renal tissues and urine together with oxidative stress and inflammation markers were examined.
    Results: Tangeretin ameliorated cisplatin-induced elevations in serum creatinine, BUN, and histopathologic changes. It also attenuated kidney oxidative stress elicited by cisplatin as demonstrated by reduced MDA and increased GSH, CAT, and SOD activities, elevated Nrf2 expression and protein levels of its downstream effectors, HO-1 and NQO-1. Tangeretin further alleviated inflammation evoked by cisplatin as indicated by reduced NF-κB p65 subunit phosphorylation with a simultaneous decrement in its downstream effectors IL-1β and TNF-α expression and protein levels. Moreover, it declined caspase-3 protein levels and TUNEL positive cells in the kidneys, the markers of apoptosis and DNA fragmentation, thus improving renal endurance. Additionally, tangeretin mitigated renal levels of KIM-1 and NGAL, as well as urinary cystatin C and β2-microglobulin concentrations, the markers of renal tubular injury.
    Conclusion: Collectively, these data signify the binary profit of tangeretin: enhancement of renal protective mechanisms against cisplatin and attenuation of renal tubular cell injuries induced by the agent.
    Keywords:  Cisplatin; KIM-1; Kidney functions; NGAL; Nephrotoxicity; Tangeretin; Tubular injury
    DOI:  https://doi.org/10.22038/ijbms.2018.32010.7691
  61. Pharmacol Rep. 2018 Dec 21. pii: S1734-1140(18)30493-6. [Epub ahead of print]71(2): 319-329
      BACKGROUND: The increased influx of free fatty acids (FFAs) into the kidney is a risk factor for diabetes nephropathy (DN). In the present study we investigated the effects of astragaloside IV (AS-IV) on FFA-induced lipid accumulation, oxidative stress, and activation of TGF-β1 signaling in human glomerular mesangial cells (HMCs).METHODS: A DN model was induced in Sprague Dawley rats by the administration of a high-fat diet and streptozocin, and HMCs were stimulated with palmitate. Lipid accumulation and FFA uptake were detected using Oil Red O and BODIPY™ FL C16 staining, respectively. The expression levels of TGF-β1, p-Smad2/3, FN, Col4 A1, NOX4, p22phox, and CD36 were evaluated by western blotting or immunofluorescence/immunohistochemistry. The level of reactive oxygen species (ROS) was detected using 2',7'-dichlorofluorescein diacetate and dihydroethidium.
    RESULTS: Exposure to palmitate induced marked lipid accumulation in HMCs, whereas co-treatment with AS-IV significantly attenuated this phenomenon. Moreover, AS-IV suppressed palmitate-induced expression of TGF-β1, p-Smad2/3, FN, Col4 A1, NOX4, and p22phox, in addition to ROS production. Notably, AS-IV reduced the palmitate-induced expression of CD36 in HMCs and DN rats. Treatment of HMCs with the CD36 inhibitor, sulfo-N-succinimidyl oleate (SSO), significantly attenuated FFA uptake, oxidative stress, and fibrosis. Nevertheless, the combined use of SSO and AS-IV did not enhance the efficacy.
    CONCLUSION: AS-IV inhibited palmitate-induced HMCs oxidative stress and fibrosis via the downregulation of CD36 expression, mediating FFA uptake and lipid accumulation.
    Keywords:  Astragaloside IV; CD36; Fibrosis; Oxidative stress; Palmitate
    DOI:  https://doi.org/10.1016/j.pharep.2018.12.008
  62. J Clin Neurosci. 2019 Feb 28. pii: S0967-5868(18)31760-0. [Epub ahead of print]
      The occurrence of intracerebral hemorrhage (ICH) costs long-standing neurologic deficits in ICH survivors, elderly ones in particular. Recent researches have proved rejuvenating effect of Growth Differentiation Factor 11 (GDF11) in improving multiple systemic diseases on old individuals. Thus, we designed this study to explore the neuroprotective effect and mechanisms of GDF11 in elderly ICH. 45 aged male Sprague-Dawley (SD) rats were randomly divided into sham + vehicle, ICH + vehicle and ICH + rGDF11 groups. ICH models were induced via injection of autologous whole blood into right basal ganglia of rats. ICH rats were given a daily injection of either recombinant (r) GDF11 at 0.1 mg/kg or vehicle for 28 days prior to operation and continued till the experiment completed. Neurological deficits, brain edema, cell apoptosis, microglial activation and heme oxygenase-1 (HO-1) positive cells were compared among each group. In addition, cytochrome c release, mitochondrial calcium buffering capacity and ATP level were monitored to explore the level of mitochondrial injury. Seen in the result, behavior disorders, severe perihematomal edema, inflammation, apoptosis, oxidative stress and mitochondria damage indicated a significant increase in ICH + vehicle group. While in ICH + rGDF11 group, administration of rGDF11 successfully reduced neurological deficits and alleviated ICH-induced edema, inflammation, apoptosis, oxidative stress, and mitochondria damage in perihematomal tissues. Collectively, our study showed that GDF11 ameliorated ICH-induced neurological deficits in elderly individuals via reducing perihematomal edema, apoptosis, inflammatory reaction, oxidative stress and improving mitochondrial dysfunction, indicating neuroprotective effect of GDF11 in elderly ICH.
    Keywords:  Apoptosis; Growth Differentiation Factor 11; Inflammation; Intracerebral hemorrhage; Mitochondria injury; Oxidative stress
    DOI:  https://doi.org/10.1016/j.jocn.2019.02.016
  63. Environ Toxicol. 2019 Mar 08.
      Osthole (Ost) is often used in treatment for cancer, inflammation and rheumatism in clinic. However, Ost-induced liver injury has been reported. In this study, we aim to investigate the possible mechanism of Ost-induced hepatotoxicity in human normal liver cells (L02). When cells were exposed to Ost, the cell viability was decreased and apoptosis rate increased, the intracellular markers of oxidative stress were changed. Simultaneously, Ost altered apoptotic related proteins levels, including Bcl-2, Bax, Cleaved-Caspase-9/-8/-3, and Pro-Caspase-3/-8. In addition, Ost enhanced the levels of endoplasmic reticulum (ER) stress proteins (GRP78/Bip, CHOP, Caspase-4, IRE1α, PERK, JNK, P-JNK, and ATF4), decreased the cell proliferation and cycle-associated protein (Phospho-Histone H3, P-Cdc25C, Cdc25C, P-Cdc2, Cdc2, and Cyclin B1) level. The results show that Ost has toxic effects on L02 cells. Furthermore, it induces apoptosis by inhibiting cell proliferation, arresting cell cycle at the G2/M phase and activating ER stress.
    Keywords:  ER stress; L02 cells; Osthole; apoptosis; cell proliferation
    DOI:  https://doi.org/10.1002/tox.22743
  64. Biochem Biophys Res Commun. 2019 Mar 01. pii: S0006-291X(19)30341-9. [Epub ahead of print]
      Several groups have demonstrated that induction of heme-oxygenase-1 (HO-1) could protect the myocardium against ischemic events; however, heme accumulation could lead to toxicity. The aim of the present study was to investigate the role of autophagy in heme toxicity. H9c2 cardiomyoblast cells were treated with different dose of hemin or cobalt-protoporphyrin IX (CoPPIX) or vehicle. Cell viability was measured by MTT assay. DCF and MitoSOX staining was employed to detect reactive oxygen species. Western blot analysis was performed to analyse the levels of HO-1, certain autophagy related proteins and pro-caspase-3 as an apoptosis marker. To study the autophagic flux, CytoID staining was carried out and cells were analyzed by fluorescence microscope and flow cytometry. Decreased cell viability was detected at high dose of hemin and CoPPIX treated H9c2 cells in a dose-dependent manner. Furthermore, at concentration of the inducers used in the present study a significantly enhanced level of ROS were detected. As it was expected both treatments induced a robust elevation of HO-1 level. In addition, the Beclin-1- independent autophagy was significantly increased, but caused a defective autophagic flux with triggered activation of caspase-3. In conclusion, these results suggest that overexpression of HO-1 by high dose of hemin and CoPPIX can induce cell toxicity in H9c2 cells via enhanced ROS level and impaired autophagy.
    Keywords:  Autophagy; Cardiomyocytes; Heme; Heme oxygenase-1; Toxicity
    DOI:  https://doi.org/10.1016/j.bbrc.2019.02.140
  65. Cell Death Dis. 2019 Mar 07. 10(3): 227
      Ionizing radiation (IR) response has been extensively investigated in BMSCs with an increasing consensus that this type of cells showed relative radiosensitivity in vitro analysis. However, the underlying mechanism of IR-induced injury of BMSCs has not been elucidated. In current study, the regulatory role of miR-22/Redd1 pathway-mediated mitochondrial reactive oxygen species (ROS) and cellular autophagy in IR-induced apoptosis of BMSCs was determined. IR facilitated the generation and accumulation of mitochondrial ROS, which promoted IR-induced apoptosis in BMSCs; meanwhile, cellular autophagy activated by IR hold a prohibitive role on the apoptosis program. The expression of miR-22 significantly increased in BMSCs after IR exposure within 24 h. Overexpression of miR-22 evidently accelerated IR-induced accumulation of mitochondrial ROS, whereas attenuated IR stimulated cellular autophagy, thus advancing cellular apoptosis. Furthermore, we verified Redd1 as a novel target for miR-22 in rat genome. Redd1 overexpression attenuated the regulatory role of miR-22 on mitochondrial ROS generation and alleviated the inhibitive role of miR-22 on cell autophagy activated by IR, thus protecting BMSCs from miR-22-mediated cell injury induced by IR exposure. These results confirmed the role of miR-22/Redd1 pathway in the regulation of IR-induced mitochondrial ROS and cellular autophagy, and subsequent cellular apoptosis.
    DOI:  https://doi.org/10.1038/s41419-019-1373-z
  66. Life Sci. 2019 Feb 27. pii: S0024-3205(19)30146-8. [Epub ahead of print]
      AIMS: The purpose of the present study was to investigate the potential antioxidant, anti-apoptotic and sperm function-preserving effects of estrogen, estrogen receptor (ER)α and ERβ agonists in a rat model of testis torsion-detorsion (T/D).MAIN METHODS: Under anesthesia, 6-8-week-old male Sprague-Dawley rats underwent sham-operation or testicular torsion by fixing left testis rotated at 720° for 2 h. After detorsion, rats were treated with ERα agonist (1 mg/kg/day, subcutaneously, sc) or ERβ agonist (1 mg/kg/day, sc) or estradiol (E2, 1 mg/kg/day, in drinking water) or vehicle on the following two days. On the third day, testicular blood-flow was recorded and then left testes were extracted for molecular and histochemical analysis.
    KEY FINDINGS: The findings showed that reduced testicular blood-flow following torsion was partially restored on the 3rd day of detorsion, while treatments with either of the ER agonists or E2 returned blood flow fully back to the control levels. When the testis-torsioned rats were given ERβ agonist during the detorsion period, tubular injury was lessened, sperm count and motility were increased, while the production of reactive oxygen metabolites and apoptosis in the testis tissues were totally suppressed. Although a down-regulated expression of androgen receptor (AR) along with a reduction in serum testosterone level was observed in the vehicle-treated T/D group, all three treatments up-regulated the expressions of AR and its mRNA, while ERα agonist and E2 suppressed the testosterone level.
    SIGNIFICANCE: ERβ receptor activation during the post-ischemic period may be beneficial in protection against torsion-related oxidant testicular injury and infertility.
    Keywords:  Apoptosis; Estrogen; Infertility; Oxidative injury; Sperm; Testicular torsion
    DOI:  https://doi.org/10.1016/j.lfs.2019.02.056
  67. Int J Mol Sci. 2019 Mar 05. pii: E1119. [Epub ahead of print]20(5):
      Wound healing implicates several biological and molecular events, such as coagulation, inflammation, migration-proliferation, and remodeling. Here, we provide an overview of the effects of malnutrition and specific nutrients on this process, focusing on the beneficial effects of curcumin. We have summarized that protein loss may negatively affect the whole immune process, while adequate intake of carbohydrates is necessary for fibroblast migration during the proliferative phase. Beyond micronutrients, arginine and glutamine, vitamin A, B, C, and D, zinc, and iron are essential for inflammatory process and synthesis of collagen. Notably, anti-inflammatory and antioxidant properties of curcumin might reduce the expression of tumor necrosis factor alpha (TNF-α) and interleukin-1 (IL-1) and restore the imbalance between reactive oxygen species (ROS) production and antioxidant activity. Since curcumin induces apoptosis of inflammatory cells during the early phase of wound healing, it could also accelerate the healing process by shortening the inflammatory phase. Moreover, curcumin might facilitate collagen synthesis, fibroblasts migration, and differentiation. Although curcumin could be considered as a wound healing agent, especially if topically administered, further research in wound patients is recommended to achieve appropriate nutritional approaches for wound management.
    Keywords:  amino-acids; curcumin; diet; macronutrients; micronutrients; minerals; nutrition; vitamins; wound; wound healing
    DOI:  https://doi.org/10.3390/ijms20051119
  68. Oral Oncol. 2019 Mar;pii: S1368-8375(19)30036-3. [Epub ahead of print]90 109-114
      OBJECTIVE: This study aimed to evaluate the effect of atorvastatin treatment on reactive oxygen species (ROS) production and tumor angiogenesis in oral squamous cell carcinomas.MATERIAL AND METHODS: An HN13 cell line was treated with 1 µM, 5 µM, and 10 µM of atorvastatin. VEGF-A gene expression was evaluated by quantitative real time PCR. VEGF-A protein expression was quantified from total protein and conditioned media by ELISA. Cellular oxidative stress was measured using 2',7'-dichlorfluorescein-diacetate (DCFH-DA). Angiogenesis assay was performed using human umbilical vein endothelial cells (HUVEC). The effect of atorvastatin on cell migration was evaluated by wound healing assay.
    RESULTS: 5 µM and 10 µM of atorvastatin significantly increased VEGF-A gene expression in the HN13 cell line. Intracellular expression of the VEGF-A protein was higher in the cells treated with 5 µM and 10 µM than in the control cells. VEGF-A protein expression was also higher in the conditioned media from the atorvastatin-treated cells than in the media from the DMSO-treated cells. 5 µM and 10 µM of atorvastatin increased oxidative stress. Regarding angiogenesis assay, 5 µM of atorvastatin resulted in higher numbers of branch points, compared to the solvent. 10 µM of atorvastatin treatment resulted in significantly reduced cell migration.
    CONCLUSIONS: This study showed that atorvastatin increases the oxidative stress and angiogenesis in oral squamous cell carcinomas. The decrease of cell migration indicates atorvastatin's inhibitory effect in oral tumors. These results suggest that atorvastatin could increase the intracellular oxidative stress in these cells, leading to a toxic microenvironment and inhibiting their metastasis.
    Keywords:  Atorvastatin; Cell migration; Oral cancer; Oxidative stress; Pathologic angiogenesis; Squamous cell carcinoma
    DOI:  https://doi.org/10.1016/j.oraloncology.2019.01.025
  69. Oxid Med Cell Longev. 2019 ;2019 8264861
      Malaria is a dangerous disease spread across several countries. Recent studies have focused on medicinal plants to discover alternative agents to the currently used drugs for malaria treatment. Here, we investigated the potential role of Indigofera oblongifolia leaf extract (IE) on hepatic inflammation in mice with Plasmodium chabaudi-infected erythrocytes. Female C57BL/6 mice were divided into three groups. The first group served as a control noninfected group, while the second and third groups were intraperitoneally injected with 106 erythrocytes parasitized by P. chabaudi. Mice from the third group were treated daily with a dose of 100 mg/kg of IE for 7 days. IE significantly reduced the number of leukocytes and apoptotic cells. The numbers of CD68-positive cells decreased in the livers of mice from the treatment group. Moreover, IE raised the hepatic antioxidant levels (glutathione and catalase) and reduced the levels of hepatic oxidative stress markers (malondialdehyde, nitric oxide, and reactive oxygen species). IE regulated some functions of the genes related to immune responses, including apoptotic genes (B-cell lymphoma-2, Bax, and caspase-3) and cytokine genes (interleukin-1β (IL-1β), IL-6, interferon-γ, and tumor necrosis factor-α). Therefore, IE exerts significant effects against malaria and protects the liver from injury caused by P. chabaudi via antioxidant and anti-inflammatory ways.
    DOI:  https://doi.org/10.1155/2019/8264861
  70. Am J Physiol Endocrinol Metab. 2019 Mar 05.
      Diabetic retinopathy (DR) is a serious diabetic complication caused by both environmental and genetic factors. Molecular mechanisms of DR may lead to the discovery of reliable prognostic indicators. The current study aimed to clarify the mechanism of microRNA-183 (miR-183) in DR in relation to the PI3K/Akt/VEGF signaling pathway . Microarray-based gene expression profiling of DR was used to identify the differentially expressed genes (DEGs). Sprague Dawley rats were used for the establishment of DR models, and then miR-183 was altered by mimic or inhibitor, or BTG1 was downregulated by siRNA to explore the regulatory mechanism of miR-183 in DR . Furthermore , the expression of miR-183, CD34, endothelial nitric oxide synthase (eNOS), BTG1 and the PI3K/Akt/VEGF signaling pathway-related genes as well as reactive oxygen species (ROS) level was determined, and the relationship between miR-183 and BTG1 was also verified. Cell growth, cell apoptosis and angiogenesis were determined. Microarray analysis revealed the involvement of miR-183 in DR via the PI3K/Akt/VEGF signaling pathway by targeting BTG1. Upregulated miR-183 and downregulated BTG1 were observed in retinal tissues of DR rats. miR-183 overexpression activated the PI3K/Akt/VEGF signaling pathway, upregulated CD34, eNOS and ROS, and inhibited BTG1. BTG1 was confirmed as a target gene of miR-183. miR-183 overexpression or BTG1 knockdown promoted cell growth and tube formation while it suppressed cell apoptosis of vascular endothelial cells in DR rats. In this study, we demonstrated that miR-183 silencing inhibiting cell growth and tube formation in vascular endothelial cells of DR rats via the PI3K/Akt/VEGF signaling pathway by upregulating BTG1.
    Keywords:  BTG1; Diabetic retinopathy; MiR-183; PI3K/Akt/VEGF signaling pathway; Vascular endothelial cell growth
    DOI:  https://doi.org/10.1152/ajpendo.00444.2018
  71. Adv Sci (Weinh). 2019 Feb 20. 6(4): 1801807
      Poor cell uptake of drugs is one of the major challenges for anticancer therapy. Moreover, the inability to release adequate drug at tumor sites and inherent multidrug resistance (MDR) may further limit the therapeutic effect. Herein, a delivery nanosystem with a charge-reversal capability and self-amplifiable drug release pattern is constructed by encapsulating β-lapachone in pH/ROS cascade-responsive polymeric prodrug micelle. The surface charge of this micellar system would be converted from negative to positive for enhanced tumor cell uptake in response to the weakly acidic tumor microenvironment. Subsequently, the cascade-responsive micellar system could be dissociated in a reactive oxygen species (ROS)-rich intracellular environment, resulting in cytoplasmic release of β-lapachone and camptothecin (CPT). Furthermore, the released β-lapachone is capable of producing ROS under the catalysis of nicotinamide adenine dinucleotide (NAD)(P)H:quinone oxidoreductase-1 (NQO1), which induces the self-amplifiable disassembly of the micelles and drug release to consume adenosine triphosphate (ATP) and downregulate P-glycoprotein (P-gp), eventually overcoming MDR. Moreover, the excessive ROS produced from β-lapachone could synergize with CPT and further propagate tumor cell apoptosis. The studies in vitro and in vivo consistently demonstrate that the combination of the pH-responsive charge-reversal, upregulation of tumoral ROS level, and self-amplifying ROS-responsive drug release achieves potent antitumor efficacy via the synergistic oxidation-chemotherapy.
    Keywords:  cascade‐response; charge‐reversal; oxidation‐chemotherapy; prodrug micelles; self‐amplifiable drug release
    DOI:  https://doi.org/10.1002/advs.201801807
  72. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2019 Jan 28. 44(1): 74-80
      In the development of oxidative stress-relevant diseases, reactive oxygen species (ROS) removal obstacle or excess production results in the damage of the body tissues and organs. Recent studies have demonstrated that nuclear factor E2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) axis played a significant role in anti-oxidative stress. The Nrf2/HO-1 axis counteracts oxidative stress injury by its resistance to inflammation, oxidation, mitochondrial damage and calcium influx, apoptosis, pyroptosis, ferroptosis and autophagy, which provides a theoretical basis for its therapeutic effect on various oxidative stress-relevant diseases in multiple organs (respiratory, cardiovascular, nervous, digestive, urinary and blood systems). Therefore, effective regulation of the Nrf2/HO-1 signal axis can be an important strategy for treatment of oxidative stress-relevant diseases.
    DOI:  https://doi.org/10.11817/j.issn.1672-7347.2019.01.012
  73. Biochim Biophys Acta Gen Subj. 2019 Feb 27. pii: S0304-4165(19)30048-0. [Epub ahead of print]1863(5): 903-916
      BACKGROUND: It has been known epidermal growth factor receptor (EGFR) frequently overexpressed in cervical cancer. High levels of EGFR expression in their tumors leads to a poor prognosis and inhibition frequently induces autophagy in cancer cells. This study aimed to investigate whether EGFR inhibition by canertinib induces autophagy and this induction influence the effect of Palladium (Pd) (II) complex and 5-fluorouracil (5-FU) especially in nontoxic doses.METHODS: Cytotoxicity was evaluated by using SRB assay. Apoptosis, autophagy, and EGFR key markers were determined by flow cytometry, fluorescence staining, and immunoblotting. Colony formation, invasion, and wound healing assays were performed to investigate cell proliferation, invasion, and migration, respectively.
    RESULTS: Blocking EGFR by the pan-ErbB tyrosine kinase inhibitor canertinib inhibited cell growth of HeLa cervical cancer cells in combination with Pd(II) complex and 5-FU. Combination of canertinib and Pd(II) complex promotes autophagy and apoptosis of HeLa cancer cells via blockade of the PI3K/AKT and MAPK/ERK pathway, which leads to cervical cancer cell death. ROS accumulation and DNA damage were increased after combinatorial treatment which causes depolarization of the mitochondrial inner membrane and leads to apoptotic cell death. Canertinib combined with Pd(II) complex leads to inhibition of migration and invasion.
    CONCLUSION: Inhibition of EGFR signaling by canertinib in combination with Pd(II) complex promotes apoptosis and autophagy via blockade of the PI3K/AKT and MAPK/ERK.
    GENERAL SIGNIFICANCE: The cytotoxic activity of Pd(II) complex and 5-FU on HeLa cells is mediated by EGFR inhibition and autophagy induction, leading to activation of mitochondrial apoptotic cell death.
    Keywords:  Apoptosis; Autophagy; Epidermal growth factor receptor; Mitochondrial depolarization; Tyrosine kinase inhibitors
    DOI:  https://doi.org/10.1016/j.bbagen.2019.02.014
  74. Front Pharmacol. 2019 ;10 115
      We previously reported that Ganoderma lucidum extract (GLE) demonstrate significant anti-cancer activity against triple negative inflammatory breast cancer models. Herein, we aimed to elucidate the bioactive compounds of GLE responsible for this anti-cancer activity. We performed NMR, X-ray crystallography and analog derivatization as well as anti-cancer activity studies to elucidate and test the compounds. We report the structures of the seven most abundant GLE compounds and their selective efficacy against triple negative (TNBC) and inflammatory breast cancers (IBC) and other human cancer cell types (solid and blood malignancies) to illustrate their potential as anti-cancer agents. Three of the seven compounds (ergosterol, 5,6-dehydroergosterol and ergosterol peroxide) exhibited significant in vitro anti-cancer activities, while we report for the first time the structure elucidation of 5,6-dehydroergosterol from Ganoderma lucidum. We also show for the first time in TNBC/IBC cells that ergosterol peroxide (EP) displays anti-proliferative effects through G1 phase cell cycle arrest, apoptosis induction via caspase 3/7 activation, and PARP cleavage. EP decreased migratory and invasive effects of cancer cells while inhibiting the expression of total AKT1, AKT2, BCL-XL, Cyclin D1 and c-Myc in the tested IBC cells. Our investigation also indicates that these compounds induce reactive oxygen species, compromising cell fate. Furthermore, we generated a superior derivative, ergosterol peroxide sulfonamide, with improved potency in IBC cells and ample therapeutic index (TI > 10) compared to normal cells. The combined studies indicate that EP from Ganoderma lucidum extract is a promising molecular scaffold for further exploration as an anti-cancer agent.
    Keywords:  EP derivatives; Ganoderma lucidum; breast cancer; ergosterol peroxide; natural product
    DOI:  https://doi.org/10.3389/fphar.2019.00115
  75. Phytother Res. 2019 Mar 05.
      The aim of this study was to examine the antitumour effects of plant phenolic acids, gallic acid (GA) and ellagic acid (EA), on human promyelocytic leukaemia sensitive HL60 cell line and its resistant sublines exhibiting two MDR phenotypes: HL60/VINC (overexpressing P-glycoprotein) and HL60/MX2 (characterized by the presence of mutated α isoform of topoisomerase II). Both studied compounds exerted comparable cytotoxic activities towards sensitive HL60 cells and their MDR counterparts. It was also found that GA and EA modulated the cellular level of reactive oxygen species in a dose-dependent and time-dependent manner. Furthermore, it was demonstrated that GA (IC90 ) and EA (IC50 and IC90 ) significantly increased the percentage of sub-G1 subpopulation of all studied leukaemia cells causing oligonucleosomal DNA fragmentation. Both compounds used at IC90 triggered mainly the apoptotic death of these cells. However, GA had no effect on the activity of caspase-3 as well as caspase-8 in sensitive HL60 cells and their MDR counterparts. In contrast, EA provoked a significant activation of these caspases in all studied leukaemia cells. It was also found that lysosomes were not involved in triggering programmed death of sensitive HL60 and MDR cells by GA and EA.
    Keywords:  ellagic acid; gallic acid; leukaemia HL60 cells; multidrug resistance; programmed cell death; reactive oxygen species
    DOI:  https://doi.org/10.1002/ptr.6317
  76. Nutrients. 2019 Mar 04. pii: E546. [Epub ahead of print]11(3):
      Retinal apoptosis plays a critical role in the progression of diabetic retinopathy (DR), a common diabetic complication. Currently, the tight control of blood glucose levels is the standard approach to prevent or delay the progression of DR. However, prevalence of DR among diabetic patients remains high. Focusing on natural nutrients or herbal medicines that can prevent or delay the onset of diabetic complications, we administered an ethanol extract of the aerial portion of Osteomeles schwerinae (OSSCE), a Chinese herbal medicine, over a period of 17 weeks to spontaneously diabetic Torii (SDT) rats. OSSCE was found to ameliorate retinal apoptosis through the regulation of advanced glycation end product (AGE) accumulation, oxidative stress, and mitochondrial function via the inhibition of NF-κB activity, in turn, through the downregulation of PKCδ, P47phox, and ERK1/2. We further demonstrated in 25 mM glucose-treated human retinal microvascular endothelial cells (HRMECs) that hyperoside (3-O-galactoside-quercetin), quercitrin (3-O-rhamnoside-quercetin), and 2″-O-acetylvitexin (8-C-(2″-O-acetyl-glucoside)-apigenin) were the active components of OSSCE that mediated its pharmacological action. Our results provide evidence that OSSCE is a powerful agent that may directly mediate a delay in the development or disease improvement in patients of DR.
    Keywords:  Osteomeles schwerinae; adjunctive effect; advanced glycation end products (AGEs); combination therapy; diabetic retinopathy (DR); human retinal microvascular endothelial cells (HRMECs); mitochondrial function; oxidative stress; retinal apoptosis; spontaneously diabetic Torii (SDT) rat
    DOI:  https://doi.org/10.3390/nu11030546
  77. Chemosphere. 2019 Feb 18. pii: S0045-6535(19)30336-4. [Epub ahead of print]224 280-288
      Although emerging evidence suggests positive association of arsenic (As) or sulfur dioxide (SO2) exposure with human diseases, reports concerning the effects of co-exposure of As and SO2 are lacking. Moreover, there is insufficient information in the literature about As and SO2 co-exposure to renal injury. In this study, we focus on the environmental problems of excessive As and SO2 that co-exist in many coal consumption areas. We used both C57BL/6 mice and 293T cells to detect toxicities of As and SO2 exposure alone or in combination. Our results showed that co-exposure significantly increased the hazard compared with exposure to As or SO2 alone. Mouse kidney tissue slices showed that co-exposure caused more severe diffuse sclerosing glomerulonephritis than As and SO2 exposure alone. Meanwhile experiments showed that apoptosis was aggravated by co-exposure of As and SO2 in 293T cells. Because As and SO2 cause cell toxicity through increasing oxidative stress, next we detected ROS and other oxidative stress parameters, and the results showed oxidative stress was increased by co-exposure compared with the other three groups. The expression levels of downstream genes in the NF-κB and caspase pathways were higher in the co-exposure group than in the groups of As or SO2 exposure alone in mice and 293T cells. Based on the above results, co-exposure could induce higher toxicity in vitro and in vivo compared with single exposure to As or SO2, indicating that people living in places that contaminated by As and SO2 may have higher chance to get renal injury.
    Keywords:  Apoptosis; As; Inflammation; ROS; SO(2)
    DOI:  https://doi.org/10.1016/j.chemosphere.2019.02.111
  78. Proc Natl Acad Sci U S A. 2019 Mar 06. pii: 201819869. [Epub ahead of print]
      Melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL-24) is a multifunctional cytokine displaying broad-spectrum anticancer activity in vitro or in vivo in preclinical animal cancer models and in a phase 1/2 clinical trial in patients with advanced cancers. mda-7/IL-24 targets specific miRNAs, including miR-221 and miR-320, for down-regulation in a cancer-selective manner. We demonstrate that mda-7/IL-24, administered through a replication incompetent type 5 adenovirus (Ad.mda-7) or with His-MDA-7/IL-24 protein, down-regulates DICER, a critical regulator in miRNA processing. This effect is specific for mature miR-221, as it does not affect Pri-miR-221 expression, and the DICER protein, as no changes occur in other miRNA processing cofactors, including DROSHA, PASHA, or Argonaute. DICER is unchanged by Ad.mda-7/IL-24 in normal immortal prostate cells, whereas Ad.mda-7 down-regulates DICER in multiple cancer cells including glioblastoma multiforme and prostate, breast, lung, and liver carcinoma cells. MDA-7/IL-24 protein down-regulates DICER expression through canonical IL-20/IL-22 receptors. Gain- and loss-of-function studies confirm that overexpression of DICER rescues deregulation of miRNAs by mda-7/IL-24, partially rescuing cancer cells from mda-7/IL-24-mediated cell death. Stable overexpression of DICER in cancer cells impedes Ad.mda-7 or His-MDA-7/IL-24 inhibition of cell growth, colony formation, PARP cleavage, and apoptosis. In addition, stable overexpression of DICER renders cancer cells more resistant to Ad.mda-7 inhibition of primary and secondary tumor growth. MDA-7/IL-24-mediated regulation of DICER is reactive oxygen species-dependent and mediated by melanogenesis-associated transcription factor. Our research uncovers a distinct role of mda-7/IL-24 in the regulation of miRNA biogenesis through alteration of the MITF-DICER pathway.
    Keywords:  DICER; MITF; ROS; mda-7/IL-24; miRNA
    DOI:  https://doi.org/10.1073/pnas.1819869116
  79. Cell Death Dis. 2019 Mar 08. 10(3): 232
      Betulinic acid (BA) and its derivatives are a class of high-profile drug candidates, but their anticancer effects on resistant cancer have rarely been reported. Although a few studies indicated mitophagy is related with drug resistance, its role in different cancer types and anticancer agents treatment remains largely unclear. Here, we find that B5G1, a new derivative of BA, induces cell death in multidrug resistant cancer cells HepG2/ADM and MCF-7/ADR through mitochondrial-apoptosis pathway. B5G1 also triggers mitophagy independent on Atg5/Beclin 1. Further mechanistic study indicates that B5G1 upregulates PTEN-induced putative kinase 1 (PINK1) to recruit Parkin to mitochondria followed by ubiquitination of Mfn2 to initiate mitophagy. Inhibition of mitophagy by PINK1 siRNA, mdivi-1, or bafilomycin A1 (Baf A1) promotes B5G1-induced cell death. In addition, ROS production and mitochondrial damage in B5G1-treated HepG2/ADM cells cause mitochondrial apoptosis and mitophagy. In vivo study shown that B5G1 dramatically inhibits HepG2/ADM xenograft growth accompanied by apoptosis and mitophagy induction. Together, our results provide the first demonstration that B5G1, as a novel mitophagy inducer, has the potential to be developed into a drug candidate for treating multidrug resistant cancer.
    DOI:  https://doi.org/10.1038/s41419-019-1470-z
  80. Int J Radiat Biol. 2019 Mar 04. 1-27
      PURPOSE: The present study aimed to investigate the potential protective effects of icariin both in vivo and in vitro, an active flavonoid glucoside derived from medicinal herb Epimedium, and its possible mechanisms against radiation-induced injury.METHODS: Male C57BL/6 mice were exposed to lethal dose (7 Gy) or sub-lethal dose (4 Gy) of whole body radiation by X-ray at a dose rate of ∼0.55 Gy/min, and icariin was given three times at 24 h and 30 min before and 24 h after the irradiation. After irradiation, hematological, biochemical, and histological evaluations were performed. We further determined the effect of icariin on radiation-induced cytotoxicity and changes in apoptosis-related protein expression.
    RESULTS: Icariin enhanced the 30-day survival rates (20 and 40 mg/kg) in a dose-dependent manner, and protected the radiosensitive organs such as intestine and testis from the radiation damages. Moreover, hematopoietic damage by radiation was significantly decreased in icariin-treated mice as demonstrated by the increases in number of peripheral blood cells, bone marrow cells (1.7-fold), and spleen colony forming units (1.7-fold). In addition, icariin decreased the radiation-induced oxidative stress by modulating endogenous antioxidant levels. Subsequent in vitro studies showed that icariin effectively increased cell viability (1.4-fold) and suppressed the expression of apoptosis-related proteins after irradiation.
    CONCLUSION: These results suggest that icariin has significant protective effects against radiation-induced damages partly through its anti-oxidative and anti-apoptotic properties.
    Keywords:  X-ray; icariin; mice; radioprotection; whole body irradiation
    DOI:  https://doi.org/10.1080/09553002.2019.1589021
  81. Vet Res. 2019 Mar 05. 50(1): 19
      Porcine circovirus type 2 (PCV2) is an economically important swine pathogen but some extra trigger factors are required for the development of PCV2-associated diseases. By evaluating cap protein expression, viral DNA copies and the number of infected cells, the present study further confirmed that oxidative stress can promote PCV2 replication. The results showed that oxidative stress induced autophagy in PCV2-infected PK15 cells. Blocking autophagy with inhibitor 3-methyladenine or ATG5-specific siRNA significantly inhibited oxidative stress-promoted PCV2 replication. Importantly, autophagy inhibition significantly increased apoptosis in oxidative stress-treated PK15 cells. Suppression of apoptosis by benzyloxycarbonyl-Val-Ala-Asp fluoromethylketone in conditions of autophagy inhibition restored PCV2 replication. Taken together, autophagy protected host cells against potential apoptosis and then contributed to PCV2 replication promotion caused by oxidative stress. Our findings can partly explain the pathogenic mechanism of PCV2 related to the oxidative stress-induced autophagy.
    DOI:  https://doi.org/10.1186/s13567-019-0637-z
  82. Biol Pharm Bull. 2019 Mar 02.
      Endoplasmic reticulum (ER) stress-mediated apoptosis pathway is considered to play a vital role in mediating stroke and other cerebrovascular diseases. Previous studies have showed that vascular endothelial growth factor (VEGF) antagonism reduced cerebral ischemic/reperfusion (CI/R) damage, but whether attenuation of ER stress-induced apoptosis is contributing to its mechanisms remains elusive. Our study aimed to investigate the protective effect of VEGF antagonism on CI/R-induced injury. First, oxygen-glucose deprivation and re-oxygenation (OGD/R) BEND3 cell model was constructed to estimate siRNA-VEGF on damage of endothelial cells. Next, in animal model, CI/R mice were induced by middle cerebral artery occlusion (MCAO) for 2 h followed by 24 h reperfusion to investigate cerebral tissue damage. For treatment group, mice received 100 µg/kg anti-VEGF antibodies at 30 min before MCAO, followed by 24 h reperfusion. Our findings demonstrated that pre-administration of siRNA-VEGF before OGD/R changed the biological characteristics of BEND3 cells, reversed the levels of X-box binding protein-1 (XBP-1) and glucose-regulated protein 78 (GRP78), showing siRNA-VEGF attenuated, at least in part, the oxidative damage in OGD/R cell by down-regulating ER stress. In mice experiment, pre-administration of anti-VEGF antibody reduced the brain infarct volume and edema extent and improved neurological scores outcome of CI/R injury mice. Pathological and TUNEL staining results also confirmed this protective effect. The expressions of VEGF, CATT/EBP homologous protein (CHOP), inositol requiring enzyme 1α (IRE-1α), and cleaved-caspase12 and jun N-terminal kinase (JNK) phosphorylation were also prominently decreased. These results suggested that inhibition of endogenous VEGF attenuates CI/R-induced injury via inhibiting ER stress-mediated apoptosis.
    Keywords:  Apoptosis; Cerebral ischemia/reperfusion; Endoplasmic reticulum stress; Vascular endothelial growth factor
    DOI:  https://doi.org/10.1248/bpb.b18-00628
  83. Benef Microbes. 2019 Mar 04. 1-12
      Neurodegeneration has been linked to changes in the gut microbiota and this study compares the neuroprotective capability of two bacterial consortia, known as Lab4 and Lab4b, using the established SH-SY5Y neuronal cell model. Firstly, varying total antioxidant capacities (TAC) were identified in the intact cells from each consortia and their secreted metabolites, referred to as conditioned media (CM). 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and Crystal Violet (CV) assays of cell viability revealed that Lab4 CM and Lab4b CM could induce similar levels of proliferation in SH-SY5Y cells and, despite divergent TAC, possessed a comparable ability to protect undifferentiated and retinoic acid-differentiated cells from the cytotoxic actions of rotenone and undifferentiated cells from the cytotoxic actions of 1-methyl-4-phenylpyridinium iodide (MPP+). Lab4 CM and Lab4b CM also had the ability to attenuate rotenone-induced apoptosis and necrosis with Lab4b inducing the greater effect. Both consortia showed an analogous ability to attenuate intracellular reactive oxygen species accumulation in SH-SY5Y cells although the differential upregulation of genes encoding glutathione reductase and superoxide dismutase by Lab4 CM and Lab4b CM, respectively, implicates the involvement of consortia-specific antioxidative mechanisms of action. This study implicates Lab4 and Lab4b as potential neuroprotective agents and justifies their inclusion in further in vivo studies.
    Keywords:  ; anti-oxidant; neurodegeneration; neurone; rotenone
    DOI:  https://doi.org/10.3920/BM2018.0105