42 research outputs found

    Antileukemic Potential of Momordica charantia Seed Extracts on Human Myeloid Leukemic HL60 Cells

    Get PDF
    Momordica charantia (bitter gourd) has been used in the traditional system of medicine for the treatment of various diseases. Anticancer activity of M. charantia extracts has been demonstrated by numerous in vitro and in vivo studies. In the present study, we investigated the differentiation inducing potential of fractionated M. charantia seed extracts in human myeloid HL60 cells. We found that the HL60 cells treated with the fractionated seed extracts differentiated into granulocytic lineage as characterized by NBT staining, CD11b expression, and specific esterase activity. The differentiation inducing principle was found to be heat-stable, and organic in nature. The differentiation was accompanied by a downregulation of c-myc transcript, indicating the involvement of c-myc pathway, at least in part, in differentiation. Taken together these results indicate that fractionated extracts of M. charantia seeds possess differentiation inducing activity and therefore can be evaluated for their potential use in differentiation therapy for leukemia in combination with other inducers of differentiation

    Quercetin 3-Glucoside Protects Neuroblastoma (SH-SY5Y) Cells in Vitro against Oxidative Damage by Inducing Sterol Regulatory Element-binding Protein-2-mediated Cholesterol Biosynthesis

    Get PDF
    The flavonoid quercetin 3-glucoside (Q3G) protected SH-SY5Y, HEK293, and MCF-7 cells against hydrogen peroxide-induced oxidative stress. cDNA microarray studies suggested that Q3G-pretreated cells subjected to oxidative stress up-regulate the expression of genes associated with lipid and cholesterol biosynthesis. Q3G pretreatment elevated both the expression and activation of sterol regulatory element-binding protein-2 (SREBP-2) only in SH-SY5Y cells subjected to oxidative stress. Inhibition of SREBP-2 expression by small interfering RNA or small molecule inhibitors of 2,3-oxidosqualene:lanosterol cyclase or HMG-CoA reductase blocked Q3G-mediated cytoprotection in SH-SY5Y cells. By contrast, Q3G did not protect either HEK293 or MCF-7 cells via this signaling pathway. Moreover, the addition of isopentenyl pyrophosphate rescued SH-SY5Y cells from the inhibitory effect of HMG-CoA reductase inhibition. Last, Q3G pretreatment enhanced the incorporation of [(14)C]acetate into [(14)C]cholesterol in SH-SY5Y cells under oxidative stress. Taken together, these studies suggest a novel mechanism for flavonoid-induced cytoprotection in SH-SY5Y cells involving SREBP-2-mediated sterol synthesis that decreases lipid peroxidation by maintaining membrane integrity in the presence of oxidative stress

    4-Hydroxynonenal Regulates Mitochondrial Function in Human Small Airway Epithelial Cells

    No full text
    Prolonged exposure to oxidative stress causes Acute Lung Injury (ALI) and significantly impairs pulmonary function. Previously we have demonstrated that mitochondrial dysfunction is a key pathological factor in hyperoxic ALI. While it is known that hyperoxia induces the production of stable, but toxic 4-hydroxynonenal (4-HNE) molecule, it is unknown how the reactive aldehyde disrupts mitochondrial function. Our previous in vivo study indicated that exposure to hyperoxia significantly increases 4-HNE-Protein adducts, as well as levels of MDA in total lung homogenates. Based on the in vivo studies, we explored the effects of 4-HNE in human small airway epithelial cells (SAECs). Human SAECs treated with 25 Ī¼M of 4-HNE showed a significant decrease in cellular viability and increased caspase-3 activity. Moreover, 4-HNE treated SAECs showed impaired mitochondrial function and energy production indicated by reduced ATP levels, mitochondrial membrane potential, and aconitase activity. This was followed by a significant decrease in mitochondrial oxygen consumption and depletion of the reserve capacity. The direct effect of 4-HNE on the mitochondrial respiratory chain was confirmed using Rotenone. Furthermore, SAECs treated with 25 Ī¼M 4-HNE showed a time-dependent depletion of total Thioredoxin (Trx) proteins and Trx activity. Taken together, our results indicate that 4-HNE induces cellular and mitochondrial dysfunction in human SAECs, leading to an impaired endogenous antioxidant response

    4-Hydroxynonenal Regulates Mitochondrial Function in Human Small Airway Epithelial Cells

    No full text
    Prolonged exposure to oxidative stress causes Acute Lung Injury (ALI) and significantly impairs pulmonary function. Previously we have demonstrated that mitochondrial dysfunction is a key pathological factor in hyperoxic ALI. While it is known that hyperoxia induces the production of stable, but toxic 4-hydroxynonenal (4-HNE) molecule, it is unknown how the reactive aldehyde disrupts mitochondrial function. Our previous in vivo study indicated that exposure to hyperoxia significantly increases 4-HNE-Protein adducts, as well as levels of MDA in total lung homogenates. Based on the in vivo studies, we explored the effects of 4-HNE in human small airway epithelial cells (SAECs). Human SAECs treated with 25 Ī¼M of 4-HNE showed a significant decrease in cellular viability and increased caspase-3 activity. Moreover, 4-HNE treated SAECs showed impaired mitochondrial function and energy production indicated by reduced ATP levels, mitochondrial membrane potential, and aconitase activity. This was followed by a significant decrease in mitochondrial oxygen consumption and depletion of the reserve capacity. The direct effect of 4-HNE on the mitochondrial respiratory chain was confirmed using Rotenone. Furthermore, SAECs treated with 25 Ī¼M 4-HNE showed a time-dependent depletion of total Thioredoxin (Trx) proteins and Trx activity. Taken together, our results indicate that 4-HNE induces cellular and mitochondrial dysfunction in human SAECs, leading to an impaired endogenous antioxidant response

    Deletion of P2X7 Attenuates Hyperoxia-induced Acute Lung Injury via Inflammasome Suppression

    No full text
    Increasing evidence shows that hyperoxia is a serious complication of oxygen therapy in acutely ill patients that causes excessive production of free radicals leading to hyperoxia-induced acute lung injury (HALI). Our previous studies have shown that P2X7 receptor activation is required for inflammasome activation during HALI. However, the role of P2X7 in HALI is unclear. The main aim of this study was to determine the effect of P2X7 receptor gene deletion on HALI. Wild-type (WT) and P2X7 knockout (P2X7 KO) mice were exposed to 100% O2 for 72 h. P2X7 KO mice treated with hyperoxia had enhanced survival in 100% O2 compared with the WT mice. Hyperoxia-induced recruitment of inflammatory cells and elevation of IL-1Ī², TNF-Ī±, monocyte chemoattractant protein-1, and IL-6 levels were attenuated in P2X7 KO mice. P2X7 deletion decreased lung edema and alveolar protein content, which are associated with enhanced alveolar fluid clearance. In addition, activation of the inflammasome was suppressed in P2X7-deficient alveolar macrophages and was associated with suppression of IL-1Ī² release. Furthermore, P2X7-deficient alveolar macrophage in type II alveolar epithelial cells (AECs) coculture model abolished protein permeability across mouse type II AEC monolayers. Deletion of P2X7 does not lead to a decrease in epithelial sodium channel expression in cocultures of alveolar macrophages and type II AECs. Taken together, these findings show that deletion of P2X7 is a protective factor and therapeutic target for the amelioration of hyperoxia-induced lung injury

    SOCS-1 rescues IL-1Ī²-mediated suppression of epithelial sodium channel in mouse lung epithelial cells via ASK-1

    No full text
    Background: Acute lung injury (ALI) is characterized by alveolar damage, increased levels of pro-inflammatory cytokines and impaired alveolar fluid clearance. Recently, we showed that the deletion of Apoptosis signal-regulating kinase 1 (ASK1) protects against hyperoxia-induced acute lung injury (HALI) by suppressing IL-1Ī² and TNF-Ī±. Previously, our data revealed that the suppressor of cytokine signaling-1 (SOCS-1) overexpression restores alveolar fluid clearance in HALI by inhibiting ASK-1 and suppressing IL-1Ī² levels. Furthermore, IL-1Ī² is known to inhibit the expression of epithelial sodium channel Ī±-subunit (ENaC) via a p38 MAPK signaling pathway. Objective: To determine whether SOCS-1 overexpression in MLE-12 cells would protect against IL-1Ī²-mediated depletion of Ī±ENaC by suppressing ASK-1 expression. Methods: We co-transfected MLE-12 cells with SOCS-1 overexpressing plasmid with or without IL-1Ī² in the presence or absence of sodium channel inhibitor, amiloride. We measured potential difference, transepithelial current, resistance, and sodium uptake levels across MLE-12 cells. We studied the effect of ASK-1 depletion, as well as ASK-1 and SOCS-1 overexpression on Ī±ENaC expression. Results: SOCS-1 overexpression sufficiently restored transepithelial current and resistance in MLE-12 cells treated with either IL-1Ī² or amiloride. The Ī±ENaC mRNA levels and sodium transport were increased in SOCS-1 overexpressing MLE-12 cells exposed to IL-1Ī². Depletion of ASK-1 in MLE-12 cells increased Ī±ENaC mRNA levels. Interestingly, SOCS-1 overexpression restored Ī±ENaC expression in MLE-12 cells in the presence of ASK-1 overexpression. Conclusion: Collectively, these findings suggest that SOCS-1 may exert its protective effect by rescuing Ī±ENaC expression via suppression of ASK-1

    SOCS-1 rescues IL-1Ī²-mediated suppression of epithelial sodium channel in mouse lung epithelial cells via ASK-1

    No full text
    Background: Acute lung injury (ALI) is characterized by alveolar damage, increased levels of pro-inflammatory cytokines and impaired alveolar fluid clearance. Recently, we showed that the deletion of Apoptosis signal-regulating kinase 1 (ASK1) protects against hyperoxia-induced acute lung injury (HALI) by suppressing IL-1Ī² and TNF-Ī±. Previously, our data revealed that the suppressor of cytokine signaling-1 (SOCS-1) overexpression restores alveolar fluid clearance in HALI by inhibiting ASK-1 and suppressing IL-1Ī² levels. Furthermore, IL-1Ī² is known to inhibit the expression of epithelial sodium channel Ī±-subunit (ENaC) via a p38 MAPK signaling pathway. Objective: To determine whether SOCS-1 overexpression in MLE-12 cells would protect against IL-1Ī²-mediated depletion of Ī±ENaC by suppressing ASK-1 expression. Methods: We co-transfected MLE-12 cells with SOCS-1 overexpressing plasmid with or without IL-1Ī² in the presence or absence of sodium channel inhibitor, amiloride. We measured potential difference, transepithelial current, resistance, and sodium uptake levels across MLE-12 cells. We studied the effect of ASK-1 depletion, as well as ASK-1 and SOCS-1 overexpression on Ī±ENaC expression. Results: SOCS-1 overexpression sufficiently restored transepithelial current and resistance in MLE-12 cells treated with either IL-1Ī² or amiloride. The Ī±ENaC mRNA levels and sodium transport were increased in SOCS-1 overexpressing MLE-12 cells exposed to IL-1Ī². Depletion of ASK-1 in MLE-12 cells increased Ī±ENaC mRNA levels. Interestingly, SOCS-1 overexpression restored Ī±ENaC expression in MLE-12 cells in the presence of ASK-1 overexpression. Conclusion: Collectively, these findings suggest that SOCS-1 may exert its protective effect by rescuing Ī±ENaC expression via suppression of ASK-1

    Testis transcript mRNP and polysome occupancy in single and compound heterozygotes.

    No full text
    <p>(A) Comparison of mutant versus wildtype mRNP or polysome abundance of expressed transcripts demonstrating global decreases in mRNP abundance and global increases in polysome abundance in compound heterozygotes but not single heterozygotes relative to wildtype. Data represented as mRNP or polysome abundance normalized by total RNA abundance and averaged across biological replicates. Red pointsā€”transcripts with significant differences between wildtype and mutant (t-test, N = 2, p-value < 0.05) within either the mRNP or polysome fractions. Yellow pointsā€”transcripts with significant differences between wildtype and mutant in both mRNP and polysome fractions. (B) Intersect of transcripts with differential mRNP or polysome occupancy in the three mutant models demonstrating limited overlap between the single and compound heterozygotes. (C) Intersect of transcripts with differential mRNP and polysome occupancy in compound heterozygotes. Transcripts within the overlap represent high confidence targets for abnormal translation repression.</p

    Translational repression and expression of high confidence targets.

    No full text
    <p>(A) Ratio of polysome to mRNP occupancy across genotypes as compared to wildtype demonstrating substantially higher polysome occupancy in compound heterozygotes when compared to single heterozygotes. Data represented as average across biological duplicates. (B) Expression of transcripts with altered mRNP and polysome occupancy compared across genotypes demonstrating minimal change in overall expression demonstrating altered polysome occupancy is not driven by differences in total expression. (C) Cell-type expression of high confidence targets demonstrating enriched expression in YBX2 and 3 positive cell types. Expressionā€”log<sub>2</sub>(average TPM). Black dotā€”median value. (D) Polysome to mRNP ratio of transcripts relevant to spermatogenesis showing increased polysome occupancy in compound heterozygotes relative to wildtype. Note the only transcript with a well-characterized YRS is <i>Spata18</i>. Data is represented as the average polysome to mRNP ratio Ā± SD. (E) Western blot detection of MAEL, NSUN2, and GAPDH in whole adult testis protein lysates from wildtype, single heterozygote, and compound heterozygous mutants demonstrating increase MAEL and NSUN2, but not GAPDH, abundance in the compound heterozygous mutant testis relative to wildtype.</p
    corecore