naphthoquinones and Hypoxia

naphthoquinones has been researched along with Hypoxia* in 12 studies

Other Studies

12 other study(ies) available for naphthoquinones and Hypoxia

ArticleYear
Plumbagin Suppresses Breast Cancer Progression by Downregulating HIF-1α Expression via a PI3K/Akt/mTOR Independent Pathway under Hypoxic Condition.
    Molecules (Basel, Switzerland), 2022, Sep-05, Volume: 27, Issue:17

    Topics: Breast Neoplasms; Cell Hypoxia; Cell Line, Tumor; Female; Humans; Hypoxia; Hypoxia-Inducible Factor 1, alpha Subunit; Naphthoquinones; Phosphatidylinositol 3-Kinases; Proto-Oncogene Proteins c-akt; TOR Serine-Threonine Kinases; Vascular Endothelial Growth Factor A

2022
Identification of Celastramycin as a Novel Therapeutic Agent for Pulmonary Arterial Hypertension.
    Circulation research, 2019, 07-19, Volume: 125, Issue:3

    Pulmonary arterial hypertension (PAH) is characterized by enhanced proliferation of pulmonary artery smooth muscle cells (PASMCs) accompanying increased production of inflammatory factors and adaptation of the mitochondrial metabolism to a hyperproliferative state. However, all the drugs in clinical use target pulmonary vascular dilatation, which may not be effective for patients with advanced PAH.. We aimed to discover a novel drug for PAH that inhibits PASMC proliferation.. We screened 5562 compounds from original library using high-throughput screening system to discover compounds which inhibit proliferation of PASMCs from patients with PAH (PAH-PASMCs). We found that celastramycin, a benzoyl pyrrole-type compound originally found in a bacteria extract, inhibited the proliferation of PAH-PASMCs in a dose-dependent manner with relatively small effects on PASMCs from healthy donors. Then, we made 25 analogs of celastramycin and selected the lead compound, which significantly inhibited cell proliferation of PAH-PASMCs and reduced cytosolic reactive oxygen species levels. Mechanistic analysis demonstrated that celastramycin reduced the protein levels of HIF-1α (hypoxia-inducible factor 1α), which impairs aerobic metabolism, and κB (nuclear factor-κB), which induces proinflammatory signals, in PAH-PASMCs, leading to reduced secretion of inflammatory cytokine. Importantly, celastramycin treatment reduced reactive oxygen species levels in PAH-PASMCs with increased protein levels of Nrf2 (nuclear factor erythroid 2-related factor 2), a master regulator of cellular response against oxidative stress. Furthermore, celastramycin treatment improved mitochondrial energy metabolism with recovered mitochondrial network formation in PAH-PASMCs. Moreover, these celastramycin-mediated effects were regulated by ZFC3H1 (zinc finger C3H1 domain-containing protein), a binding partner of celastramycin. Finally, celastramycin treatment ameliorated pulmonary hypertension in 3 experimental animal models, accompanied by reduced inflammatory changes in the lungs.. These results indicate that celastramycin ameliorates pulmonary hypertension, reducing excessive proliferation of PAH-PASMCs with less inflammation and reactive oxygen species levels, and recovered mitochondrial energy metabolism. Thus, celastramycin is a novel drug for PAH that targets antiproliferative effects on PAH-PASMCs.

    Topics: Animals; Cells, Cultured; Cytokines; Disease Models, Animal; Drug Evaluation, Preclinical; Energy Metabolism; High-Throughput Screening Assays; Humans; Hypoxia; Hypoxia-Inducible Factor 1, alpha Subunit; Indoles; Male; Metabolome; Mice; Mitochondria; Monocrotaline; Muscle, Smooth, Vascular; Myocytes, Smooth Muscle; Naphthoquinones; NF-E2-Related Factor 2; NF-kappa B; Oxidative Stress; Pulmonary Arterial Hypertension; Pulmonary Artery; Pyrroles; Rats; Reactive Oxygen Species; Resorcinols; Transcription Factors

2019
Shikonin protects H9C2 cardiomyocytes against hypoxia/reoxygenation injury through activation of PI3K/Akt signaling pathway.
    Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 2018, Volume: 104

    Myocardial ischemic/reperfusion (I/R) injury often leads to irreversible myocardial cell death and even heart failure, with limited therapeutic possibilities. In the present study, we evaluated the protective effects of shikonin (SHK) against hypoxia/reoxygenation (H/R)-induced cardiomyocyte damage and explored the underlying mechanisms. H9C2 cardiomyocytes were pretreated with different doses of SHK prior to H/R exposure. We observed that SHK pretreatment significantly increased cell viability, attenuated LDH release, and suppressed cardiomyocyte apoptosis induced by H/R exposure. SHK pretreatment also restored the loss of mitochondrial membrane potential (MMP) and cytochrome c release. In addition, SHK significantly enhanced the phosphorylation of Akt and GSK-3β in H/R-treated H9C2 cells. These protective effects of SHK were partially reversed by LY294002, a specific PI3K/Akt inhibitor. Therefore, our findings suggested that SHK might be a promising agent for myocardial I/R injury, and PI3K/Akt signaling plays a crucial role during this process.

    Topics: Animals; Apoptosis; Cardiotonic Agents; Cell Line; Cell Survival; Chromones; Glycogen Synthase Kinase 3 beta; Hypoxia; Membrane Potential, Mitochondrial; Mitochondria; Morpholines; Myocardial Reperfusion Injury; Myocardium; Myocytes, Cardiac; Naphthoquinones; Phosphatidylinositol 3-Kinases; Phosphorylation; Protective Agents; Proto-Oncogene Proteins c-akt; Rats; Signal Transduction

2018
Shikonin‑mediated inhibition of nestin affects hypoxia‑induced proliferation of pulmonary artery smooth muscle cells.
    Molecular medicine reports, 2018, Volume: 18, Issue:3

    The imbalance between the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) is of importance in pulmonary vascular remodeling. Shikonin, a naphthoquinone compound extracted from the Chinese medicinal herb Lithospermum erythrorhizon, inhibits the proliferation of rat smooth muscle cells (SMCs). The present study was designed to investigate the effects of shikonin on the proliferation of rat PASMCs and the possible mechanisms involved. Rat PASMCs were cultured under the following five treatment conditions: Normal control; hypoxia for 24 h; hypoxia + 1 µM shikonin for 24 h; hypoxia + 2 µM shikonin for 24 h; and hypoxia + 4 µM shikonin for 24 h. The viability of PASMCs was measured using the Cell Counting Kit‑8 assay, the mRNA expression of nestin (NES) in each group was measured by reverse transcription‑polymerase chain reaction and the protein expression of NES was measured by western blotting. The proliferation of hypoxic PASMCs transfected with NES‑specific small interfering (si)RNA decreased compared with the non‑transfected group. These results indicated that hypoxia induced the proliferation of PASMCs through the enhancement of NES expression. The treatment of hypoxic PASMCs with shikonin resulted in a significant downregulation of NES expression and the inhibition of PASMC proliferation.

    Topics: Animals; Cell Hypoxia; Cell Proliferation; Cells, Cultured; Gene Expression Regulation; Hypoxia; Male; Muscle, Smooth, Vascular; Myocytes, Smooth Muscle; Naphthoquinones; Nestin; Pulmonary Artery; Rats; RNA, Small Interfering

2018
YM155, a selective survivin inhibitor, reverses chronic hypoxic pulmonary hypertension in rats via upregulating voltage-gated potassium channels.
    Clinical and experimental hypertension (New York, N.Y. : 1993), 2015, Volume: 37, Issue:5

    To test the hypothesis that chronic hypoxic pulmonary hypertension (CH-PH) is associated with increased survivin and decreased voltage-gated potassium (KV) channels expression in pulmonary arteries, rats were randomized as: normoxia (N); normoxia + YM155, survivin suppressor (NY); hypoxia (H); hypoxia + YM155 (HY). HY group had significantly reduced pulmonary arterial pressure, right ventricular weight and right ventricular hypertrophy compared with H group. Survivin mRNA and protein were detected in pulmonary arteries of rats with CH-PH, but not rats without CH-PH. YM155 downregulated survivin protein and mRNA. KV channel expression and activity were upregulated after YM155 treatment. Survivin may play a role in the pathogenesis of CH-PH.

    Topics: Animals; Chronic Disease; Disease Models, Animal; Gene Expression Regulation; Hypertension, Pulmonary; Hypoxia; Imidazoles; Male; Microtubule-Associated Proteins; Muscle, Smooth, Vascular; Naphthoquinones; Patch-Clamp Techniques; Potassium Channels, Voltage-Gated; Pulmonary Wedge Pressure; Rats; Rats, Wistar; Real-Time Polymerase Chain Reaction; RNA; Survivin

2015
Plumbagin, a novel Nrf2/ARE activator, protects against cerebral ischemia.
    Journal of neurochemistry, 2010, Volume: 112, Issue:5

    Many phytochemicals function as noxious agents that protect plants against insects and other damaging organisms. However, at subtoxic doses, the same phytochemicals may activate adaptive cellular stress response pathways that can protect cells against a variety of adverse conditions. We screened a panel of botanical pesticides using cultured human and rodent neuronal cell models, and identified plumbagin as a novel potent activator of the nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. In vitro, plumbagin increases nuclear localization and transcriptional activity of Nrf2, and induces the expression of the Nrf2/ARE-dependent genes, such as heme oxygenase 1 in human neuroblastoma cells. Plumbagin specifically activates the Nrf2/ARE pathway in primary mixed cultures from ARE-human placental alkaline phosphatase reporter mice. Exposure of neuroblastoma cells and primary cortical neurons to plumbagin provides protection against subsequent oxidative and metabolic insults. The neuroprotective effects of plumbagin are abolished by RNA interference-mediated knockdown of Nrf2 expression. In vivo, administration of plumbagin significantly reduces the amount of brain damage and ameliorates-associated neurological deficits in a mouse model of focal ischemic stroke. Our findings establish precedence for the identification and characterization of neuroprotective phytochemicals based upon their ability to activate adaptive cellular stress response pathways.

    Topics: Animals; Cell Line, Tumor; Cell Survival; Cells, Cultured; Cerebral Cortex; Cerebral Infarction; Disease Models, Animal; Embryo, Mammalian; Gene Expression Regulation; Glucose; Heme Oxygenase-1; Humans; Hypoxia; Infarction, Middle Cerebral Artery; Mice; Mice, Inbred C57BL; Naphthoquinones; Neuroblastoma; Neurologic Examination; Neurons; Neuroprotective Agents; NF-E2-Related Factor 2; Oxidative Stress; Rats; Rats, Sprague-Dawley; Transcription Factor AP-1; Transfection

2010
Beta-carotene breakdown products enhance genotoxic effects of oxidative stress in primary rat hepatocytes.
    Carcinogenesis, 2006, Volume: 27, Issue:6

    Since it has to be expected that individuals exposed to oxidative stress who take supplements of beta-carotene are simultaneously exposed to both beta-carotene cleavage products (CPs) and oxidative stress, and both exposures have been demonstrated to cause genotoxic effects in primary rat hepatocytes, cyto- and genotoxic effects on primary rat hepatocytes after supplementation of the medium with increasing concentrations of a CP mixture during exposure to oxidative stress by treatment with either DMNQ (2,3-dimethoxy-1,4-naphthoquinone) or hypoxia/reoxygenation (Hy/Reox) was investigated. The cytological endpoints analysed were the mitotic indices, the percentages of apoptotic and necrotic cells, the percentages of micronucleated (MN) cells and the number of chromosomal aberrations (CAs) and sister chromatid exchanges (SCE). The results obtained clearly demonstrate that the CP mixture enhances the genotoxic effects of oxidative stress exposure, whereas it had no effect at all on the endpoints of cytotoxicity studied. These results further support the hypothesis that CP might be responsible for the reported carcinogenic response in the beta-CArotene and Retinol Efficacy Trial (CARET) and Alpha-Tocopherol Beta-carotene Cancer prevention (ATBC) chemoprevention trials.

    Topics: Animals; beta Carotene; Chromosome Aberrations; DNA Damage; Dose-Response Relationship, Drug; Female; Hepatocytes; Hypoxia; Metaphase; Naphthoquinones; Oxidative Stress; Oxygen; Rats; Rats, Inbred F344

2006
NO restores HIF-1alpha hydroxylation during hypoxia: role of reactive oxygen species.
    Free radical biology & medicine, 2005, Oct-01, Volume: 39, Issue:7

    The activity of hypoxia-inducible factor 1 (HIF-1) is primarily determined by stability regulation of its alpha subunit, which is stabilized under hypoxia but degraded during normoxia. Hydroxylation of HIF-1alpha by prolyl hydroxylases (PHDs) recruits the von Hippel-Lindau (pVHL) E3 ubiquitin ligase complex to initiate proteolytic destruction of the alpha subunit. Hypoxic stabilization of HIF-1alpha has been reported to be antagonized by nitric oxide (NO). By using a HIF-1alpha-pVHL binding assay, we show that NO released from DETA-NO restored prolyl hydroxylase activity under hypoxia. Destabilization of HIF-1alpha by DETA-NO was reversed by free radical scavengers such as NAC and Tiron, thus pointing to the involvement of reactive oxygen species (ROS). Therefore, we examined the effects of ROS on HIF-1alpha stabilization. Treatment of cells under hypoxia with low concentrations of the superoxide generator 2,3-dimethoxy-1,4-naphthoquinone lowered HIF-1alpha protein stabilization. In vitro HIF-1alpha-pVHL interaction assays demonstrated that low-level ROS formation increased prolyl hydroxylase activity, an effect antagonized by ROS scavengers. While determining intracellular ROS formation we noticed that reduced ROS production under hypoxia was restored by the addition of DETA-NO. We propose that an increase in ROS formation contributes to HIF-1alpha destabilization by NO donors under hypoxia via modulation of PHD activity.

    Topics: 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt; Acetylcysteine; Carcinoma, Hepatocellular; Cell Line; Humans; Hydroxylation; Hypoxia; Naphthoquinones; Nitric Oxide; Nitric Oxide Donors; Procollagen-Proline Dioxygenase; Reactive Oxygen Species; Triazenes; Tumor Cells, Cultured; Von Hippel-Lindau Tumor Suppressor Protein

2005
Pharmacological and biological evaluation of a series of substituted 1,4-naphthoquinone bioreductive drugs.
    Biochemical pharmacology, 2004, Dec-01, Volume: 68, Issue:11

    The indolequinone compound EO9 has good pharmacodynamic properties in terms of bioreductive activation and selectivity for either NAD(P)H:quinone oxidoreductase-1 (NQO1)-rich aerobic or NQO1-deficient hypoxic cells. However, its pharmacokinetic properties are poor and this fact is believed to be a major reason for EO9's lack of clinical efficacy. The purpose of this study was to develop quinone-based bioreductive drugs that retained EO9's good properties, in terms of bioreductive activation, but have improved pharmacokinetic properties. Out of 11 naphthoquinone compounds evaluated, 2-aziridinyl-5-hydroxy-1,4-naphthoquinone (compound 2), 2,3-bis(aziridinyl)-5-hydroxy-1,4-naphthoquinone (compound 3), and 2-aziridinyl-6-hydroxymethyl-1,4-naphthoquinone (compound 11) were selected for further evaluation based on good substrate specificity for NQO1 and selectivity towards NQO1-rich cells in vitro. Compound 3 was of particular interest as it also demonstrated selectivity for NQO1-rich cells under hypoxic conditions. Compound 3 was not metabolised by murine whole blood in vitro (in contrast to compounds 2, 11 and EO9) and pharmacokinetic studies in non-tumour-bearing mice in vivo (at the maximum soluble dose of 60 mg kg(-1) administered intraperitoneally) demonstrated significant improvements in plasma half-life (16.2 min) and AUC values (22.5 microM h) compared to EO9 (T(1/2) = 1.8 min, AUC = 0.184 microM h). Compound 3 also demonstrated significant anti-tumour activity against H460 and HCT-116 human tumour xenografts in vivo, whereas EO9 was inactive against these tumours. In conclusion, compound 3 is a promising lead compound that may target both aerobic and hypoxic fractions of NQO1-rich tumours and further studies to elucidate its mechanism of action and improve solubility are warranted.

    Topics: Animals; Antineoplastic Agents; Aziridines; Disease Models, Animal; Drug Screening Assays, Antitumor; Drug Stability; Female; Humans; Hypoxia; Indolequinones; Mice; NAD(P)H Dehydrogenase (Quinone); Naphthoquinones; Neoplasm Transplantation; Neoplasms, Experimental; Substrate Specificity; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2004
Oxidative stress in cultured cerebral endothelial cells induces chromosomal aberrations, micronuclei, and apoptosis.
    Journal of neuroscience research, 2003, May-01, Volume: 72, Issue:3

    There is evidence accumulating that brain microvasculature is involved critically in oxidative stress-mediated brain damage. Cultured cerebral microvascular endothelial cells were used to demonstrate the cytotoxic and genotoxic effects elicited by hypoxia/reoxygenation and DMNQ treatment in vitro. In addition, the effect of glucose deprivation during oxidative insult was assessed. The parameters determined were: 1) chromosomal aberrations; 2) induction of micronuclei; and 3) apoptosis. Our results indicate that both the exposure of the cerebral endothelial cells to 24 hr of hypoxia followed by 4 hr of reoxygenation, and treatment with the redox cycling quinone DMNQ, increased markedly the occurrence of chromosomal aberrations and micronuclei. It was found that expression of p53 was induced by oxidative stress, particularly when glucose had been omitted from the culture medium. Aglycemic culture conditions in general exacerbated the cytotoxic effects of oxidative insults, as evidenced by the increase in apoptotic cells and the decrease in the mitotic index. Interestingly, neither an elevation of cell lysis nor an increase in necrosis has been observed during our experiments. In summary, our data indicate that oxidative stress exerts considerable genotoxic and cytotoxic effects on cerebral endothelial cells, which might contribute to the progression of tissue damage in the central nervous system.

    Topics: Animals; Apoptosis; Blotting, Western; Cells, Cultured; Chromosome Aberrations; Endothelium, Vascular; Glucose; Hypoxia; L-Lactate Dehydrogenase; Micronuclei, Chromosome-Defective; Naphthoquinones; Oxidative Stress; Rats; Reperfusion Injury; Swine; Telencephalon; Time Factors; Tumor Suppressor Protein p53

2003
Inhibitory effect of a naphthazarin derivative, S64, on heat shock factor (Hsf) activation and glutathione status following hypoxia.
    Cell biology and toxicology, 2003, Volume: 19, Issue:5

    The presence of hypoxic cells in solid tumors has long been considered a problem in cancer treatment. Resistance of hypoxic cells to ionizing radiation and anticancer drugs has in part been attributed to changes in altered gene expression by hypoxia. We previously reported an activation of heat shock factor (Hsf) in murine tumor RIF cells following hypoxia and suggested that a subsequent accumulation of heat shock protein(s) (Hsp) is likely to contribute to the malignant progression of hypoxic tumor cells (Baek et al., 2001). In this study, we showed that hypoxia induced a DNA-binding activity of Hsf and activation of hsp70 gene expression in colon cancer Clone A cells, and that a naphthazarin derivative, S64, significantly inhibited the hypoxia-inducible hsp70 gene expression in Clone A cells. We also showed that S64 significantly reduced the cellular glutathione levels in this cell line. Considering the proposed effects of Hsp and glutathione on radiation and chemotherapy sensitivity, we suggest that the inhibitory effects of S64 on Hsf activation and cellular glutathione levels have potentially important clinical implications. We believe that the previously reported in vitro and in vivo anti-tumor effect of S64 (Song et al., 2000a, 2001) might be attributed, at least in part, to its effect on Hsf activation and/or glutathione depletion. We also believe that the detailed molecular mechanisms underlying the effects of S64 on Hsf and glutathione level following hypoxia deserve a more rigorous future study, the results of which could offer novel strategy to manipulate the resistance mechanisms of solid tumors.

    Topics: Animals; Antineoplastic Agents; Blotting, Western; Cell Line, Tumor; Cell Nucleus; DNA; Glutamate-Cysteine Ligase; Glutathione; HSP70 Heat-Shock Proteins; Hypoxia; Mice; Naphthoquinones; Protein Binding; Reactive Oxygen Species; Reverse Transcriptase Polymerase Chain Reaction; RNA; Time Factors

2003
Pathologic physiology and chemotherapy of Plasmodium berghei. X. Pulmonary edema and naphthoquinones.
    Experimental parasitology, 1969, Volume: 26, Issue:3

    Topics: Animals; Body Weight; Carbon Dioxide; Chemical Phenomena; Chemistry; Epinephrine; Hypoxia; Malaria; Methods; Mice; Morphine; Naphthoquinones; Organ Size; Oxygen Consumption; Plasmodium; Pulmonary Edema; Time Factors

1969