bims-noxint Biomed News
on NADPH oxidases in tumorigenesis
Issue of 2020‒01‒26
nine papers selected by
Laia Caja Puigsubira
Uppsala University


  1. Antioxid Redox Signal. 2020 Jan 22.
      SIGNIFICANCE: Despite their intrinsic cytotoxic properties, mounting evidence indicate that ROS physiologically produced by the NADPH oxidases of epithelial cells (NOX1, DUOX2) and phagocytes (NOX2) are critical for innate immune response and homeostasis of the intestinal mucosa. However, dysregulated ROS production could be a driving factor in inflammatory bowel diseases (IBD). Recent advances: In addition to NOX2, recent studies have demonstrated that NOX1- and DUOX2-derived ROS can regulate intestinal innate immune defense and homeostasis by impacting many processes, including bacterial virulence, expression of bacteriostatic proteins, epithelial renewal and restitution, and microbiota composition. Moreover, the antibacterial role of DUOX2 is a function conserved in evolution as it has been described in invertebrates, and lower and higher vertebrates. In humans, variants of the NOX2, NOX1 and DUOX2 genes, which are associated with impaired ROS production, have been identified in very early-onset IBD, but overexpression of NOX/DUOX, especially DUOX2, has also been described in IBD, suggesting that loss-of-function or excessive activity of the ROS-generating enzymes could both contribute to disease progression.CRITICAL ISSUES: Therapeutic perspectives aiming at targeting NOX/DUOX in IBD should take into account the two sides of NOX/DUOX-derived ROS in intestinal inflammation. Hence, NOX /DUOX inhibitors or ROS inducers should be considered as a function of the disease context.
    FUTURE DIRECTIONS: A thorough understanding of the physiological and pathological regulation of NOX/DUOX in the gastrointestinal tract is an absolute pre-requisite for the development of therapeutic strategies that can modulate ROS levels in space and time.
    DOI:  https://doi.org/10.1089/ars.2020.8018
  2. Biochem Biophys Res Commun. 2020 Jan 20. pii: S0006-291X(20)30007-3. [Epub ahead of print]
      Optimal levels of reactive oxygen species (ROS) play a critical role in cellular physiological function. For production of intracellular superoxide, NADPH oxidase is one of the sources. Rac1/2 and RhoA GTPases are involved in regulation of NADPH oxidase activity and Tyr42 phosphorylation of RhoA (p-Tyr42 RhoA) seems significant in this regard as it was recently shown that hydrogen peroxide was able to increase p-Tyr42 RhoA levels. Phorbol myristate acetate (PMA), a tumor promoter, also induces production of superoxides; PMA activates Src, a tyrosine kinase, and increases p-Tyr42 RhoA levels. In exploring the mechanism of PMA effects, we reduced RhoA levels in test cells with si-RhoA and then restoration of various versions of RhoA for effect in response of the cells to PMA and producing superoxides. Restoration of RhoA Y42F (a dephospho-mimic form) still had reduced superoxide formation in response to PMA, compared with WT and Y42E RhoA. This was similarly seen with assays for cell migration and proliferation with cells responding to PMA. Y27632, a ROCK (Rho associated coiled coil kinase) inhibitor, also inhibited superoxide production, and also reduced p-Y416 Src and p-p47phox levels. A ROCK active fragment was also able to phosphorylate p47phox at Ser345 residue (p-Ser345 p47phox), a component of NADPH oxidase. Overall, we demonstrate that p-Tyr42 RhoA levels increase following PMA treatment and this is through production of superoxide and activation of Src. These in turn amplify superoxide production through ROCK phophorylation of p47phox and maintain a positive feedback loop for superoxide generation, and contribute to tumor progression.
    Keywords:  PMA; ROCK; Superoxide; p-Tyr42 RhoA; p47phox
    DOI:  https://doi.org/10.1016/j.bbrc.2020.01.001
  3. J Leukoc Biol. 2020 Jan 22.
      Activation of the phagocyte NADPH oxidase involves a conformational change in Nox2. The effector in this process is p67phox and there is evidence for a change in the configuration of p67phox being required for binding to Nox2. To study this, we measured binding of p67phox to a library of Nox2 peptides and binding of NusA-Nox2 fusion proteins to p67phox . We found, serendipitously, that deletion of residues 259-279 in p67phox (p67phox Δ(259-279)), endowed it with the ability to bind selectively to Nox2 peptide 369-383 (peptide 28). There was no binding to scrambled Nox2 peptide 28 and to Nox4 peptide 28. Binding was cysteine independent and resistant to reducing and alkylating agents. Truncations of peptide 28 revealed that the actual binding site consisted of residues 375-383. Binding of p67phox Δ(259-279) to peptide 28 was mimicked by that of a (p67phox -RacGTP) chimera. Both p67phox Δ(259-279) and the (p67pho -RacGTP) chimera bound a NusA-Nox2 fusion protein, comprising residues 375-383. Specific single residue deletion mutants, within the p67phox sequence 259-279, were also bound to Nox2 peptide 28. Peptides synthesized to correspond to the 259-279 sequence in p67phox , were found to autobind p67phox , suggesting that an intramolecular bond exists in p67phox , one pole of which was located within residues 259-279. We conclude that "resting" p67phox exists in a "closed" conformation, generated by an intramolecular bond. Deletion of specific residues within the 259-279 sequence, in vitro, or interaction with RacGTP, in vivo, causes "opening" of the bond and results in binding of p67phox to a specific, previously unknown, site in Nox2.
    Keywords:  (p67phox-Rac) chimera; NADPH oxidase; NusA-Nox2 fusion protein; intramolecular bond; peptide-protein interaction; synthetic peptides
    DOI:  https://doi.org/10.1002/JLB.4A1219-607R
  4. Antioxid Redox Signal. 2020 Jan 22.
      SIGNIFICANCE: The growing incidence of neurodegenerative diseases significantly impacts the individuals who suffer from these disorders and is a major health concern globally. Although the specific mechanisms of neurodegenerative diseases are still far from being acknowledged, it is becoming clear that oxidative stress and neuroinflammation are critical contributing factors to the progression of neurodegeneration. Thus, it is conceivable that the inhibition of oxidative stress and neuroinflammation may represent promising therapeutic targets for the treatment of neurodegenerative diseases. Recent Advances: Recently, the strategy for neurodegenerative disease therapy has shifted from the use of antioxidants and conventional anti-inflammatory targets to upstream mediators due to the failure of most antioxidants and nonsteroidal anti-inflammatory drugs (NSAIDs) in clinical trials. NADPH oxidases (NOXs), a family of superoxide-producing enzyme complexes, have been identified as an upstream factor that controls both oxidative stress and neuroinflammation. Genetic inactivation or pharmacological inhibition of NOX enzymes displays potent neuroprotective effects in a broad spectrum of neurodegenerative disease models.CRITICAL ISSUES: The detailed mechanisms of how NOX enzymes regulate oxidative stress and neuroinflammation still remain unclear. Moreover, the currently available inhibitors of NOX enzymes exhibit nonspecificity, off-target effects, unsuitable pharmacokinetic properties and even high toxicity, markedly limiting their potential clinical applications.
    FUTURE DIRECTIONS: This review provides novel insights into the roles of NOXs in neurodegenerative pharmacology and indicates the types NOX enzyme inhibitors that should be identified and developed as candidates for future applications, which might reveal novel neurodegenerative disease therapies based on NOXs.
    DOI:  https://doi.org/10.1089/ars.2019.8014
  5. Nitric Oxide. 2020 Jan 20. pii: S1089-8603(19)30259-9. [Epub ahead of print]
      The metabolic disorders in diabetes, which are usually accompanied by oxidative stress and impaired nitric oxide (NO) bioavailability, increase the risk of detrimental cardiovascular complications. Herein, we investigated the therapeutic potential of dietary nitrate, which is found in high content in green leafy vegetables, on vascular oxidative stress and endothelial dysfunction in diabetic mice induced by high-fat diet and streptozotocin injection. Dietary nitrate in drinking water fuelled a nitrate-nitrite-NO pathway, which inhibited vascular oxidative stress, endothelial dysfunction and many features of metabolic syndrome in diabetic mice. These beneficial effects of nitrate on diabetic mice were abolished by PTIO (NO scavenger) treatment and significantly prevented by febuxostat (xanthine oxidoreductase inhibitor), demonstrating the central importance of NO in bioactivation of nitrate. The favorable effects of nitrate were not further influenced by apocynin (NADPH oxidase inhibitor), suggesting NADPH oxidase as a possible target. In high glucose-incubated vascular endothelial cells, NO donor attenuated oxidative stress and endothelial dysfunction via the inhibition of NADPH oxidase, where a heme oxygenase-1 (HO-1)-dependent mechanism was demonstrated for the antioxidant abilities of NO. Altogether, boosting this nitrate-nitrite-NO signaling pathway resulted in the decreases of NADPH oxidase-derived oxidative stress, endothelial dysfunction and metabolic disorders in diabetic vasculature. These findings may have novel implications for the preventive strategy against diabetes-induced vascular dysfunction and associated complications.
    Keywords:  Diabetes; Endothelial dysfunction; NADPH oxidase; Nitrate; Nitric oxide
    DOI:  https://doi.org/10.1016/j.niox.2020.01.007
  6. Oxid Med Cell Longev. 2019 ;2019 9736047
      Objective: Glaucoma is a leading cause of severe visual impairment and blindness. Although high intraocular pressure (IOP) is an established risk factor for the disease, the role of abnormal ocular vessel function in the pathophysiology of glaucoma gains more and more attention. We tested the hypothesis that elevated intraocular pressure (IOP) causes vascular dysfunction in the retina.Methods: High IOP was induced in one group of mice by unilateral cauterization of three episcleral veins. The other group received sham surgery only. Two weeks later, retinal vascular preparations were studied by video microscopy in vitro. Reactive oxygen species (ROS) levels and expression of hypoxia markers and of prooxidant and antioxidant redox genes as well as of inflammatory cytokines were determined.
    Results: Strikingly, responses of retinal arterioles to stepwise elevation of perfusion pressure were impaired in the high-IOP group. Moreover, vasodilation responses to the endothelium-dependent vasodilator, acetylcholine, were markedly reduced in mice with elevated IOP, while no differences were seen in response to the endothelium-independent nitric oxide donor, sodium nitroprusside. Remarkably, ROS levels were increased in the retinal ganglion cell layer including blood vessels. Expression of the NADPH oxidase isoform, NOX2, and of the inflammatory cytokine, TNF-α, was increased at the mRNA level in retinal explants. Expression of NOX2, but not of the hypoxic markers, HIF-1α and VEGF-A, was increased in the retinal ganglion cell layer and in retinal blood vessels at the protein level.
    Conclusion: Our data provide first-time evidence that IOP elevation impairs autoregulation and induces endothelial dysfunction in mouse retinal arterioles. Oxidative stress and inflammation, but not hypoxia, appear to be involved in this process.
    DOI:  https://doi.org/10.1155/2019/9736047
  7. J Immunol. 2020 Jan 24. pii: ji1900551. [Epub ahead of print]
      LETM1 domain-containing protein 1 (LETMD1), also known as HCCR-1, is a mitochondrial protein and is known to regulate p53 and STAT3 activities in cancer cells. In this study, we present, for the first time (to our knowledge), data indicating that LETMD1 suppresses multiple immune responses in monocyte/macrophage lineage cells and mouse primary macrophages. Attenuation of LETMD1 expression with specific small interfering RNA and short hairpin RNA constructs enhanced LPS-induced expressions of inflammatory mediators in macrophages. In addition, LETMD1 attenuation caused potentiation of phagocytosis as well as migration in a macrophage-like cell line, U937. These enhancing effects were associated with altered activation of signaling adaptors (such as NF-κB, MAPKs, p53, and JAK-STAT) involved in TLR4 signaling. Especially, LETMD1 selectively regulated TLR4-induced NF-κB activation via MyD88 but not via TIR-domain-containing adapter-inducing IFN-β (TRIF). Attenuation of LETMD1 expression caused mitochondrial hyperpolarization and subsequent decrease in ATP production and increase in mitochondrial/cellular reactive oxygen species (ROS) and intracellular calcium levels. LETMD1 attenuation also enhanced LPS-induced expression of NADPH oxidase (NOX) 2, the main producer of cellular ROS in phagocytes, through augmenting IFN regulatory factor 1. Accordingly, treatment with ROS scavenger, NOX2 suppressing agents, or calcium chelators resulted in suppression of LPS-induced cytokine production as well as NF-κB activation in cells with LETMD1 attenuation. These findings reveal a previously unknown function of LETMD1 and provide evidences showing LETMD1 negatively regulates macrophage functions by modulating mitochondrial function, subsequent ROS generation, and NF-κB activation.
    DOI:  https://doi.org/10.4049/jimmunol.1900551
  8. Redox Biol. 2020 Jan 14. pii: S2213-2317(19)31116-4. [Epub ahead of print]30 101433
      Studies revealed that the use of renin-angiotensin-aldosterone system antagonism is not associated with a statistically significant reduction in the risk of cardiovascular events in patients with chronic kidney disease (CKD) compared with that in the general population. We tested the hypothesis that indoxyl sulfate (IS) can interfere with the protective effect of valsartan-mediated on endothelial function in vitro and neovascularization in mice underwent subtotal nephrectomy. In human aortic endothelial cells, we first demonstrated that IS impaired the valsartan-mediated phosphorylation of eNOSThr495, nitric oxide production and tube formation via NADPH oxidase (NOX) and protein kinase C (PKC) phosphorylation, but this effect was suppressed by cotreatment with apocynin and calphostin C. In vivo, IS attenuated valsartan-induced angiogenesis in Matrigel plugs in mice. Moreover, in subtotal nephrectomy mice who underwent hindlimb ischemic surgery, valsartan significantly increased the mobilization of endothelial progenitor cells in circulation as well as the reperfusion of blood flow and density of CD31+ capillaries in ischemic limbs. However, IS attenuated the protective effect of valsartan-induced neovascularization and increased the expression of p-PKCαSer657 and p-eNOSThr497 in ischemic limbs. Cotreatment of apocynin and calphostin C reversed the IS impaired-neovascularization and decreased the expression of p-PKCαSer657 and p-eNOSThr497 in ischemic limbs. Our study suggests that the NOX/PKC/eNOS signaling pathway plays a pivotal role in the IS-mediated inhibition of valsartan-conferred beneficial effects on endothelial function in vitro and neovascularization in subtotal nephrectomy mice. We proposed a novel causative role for IS in cardiovascular complications in CKD patients.
    Keywords:  Chronic kidney disease; Indoxyl sulfate; Neovascularization; Valsartan
    DOI:  https://doi.org/10.1016/j.redox.2020.101433
  9. Arch Biochem Biophys. 2020 Jan 18. pii: S0003-9861(19)31060-4. [Epub ahead of print] 108275
      Stroke is one of the world's most deadly pathologies, and the rate of stroke recurrence is high. However, due to the complex nature of ischemia and reperfusion injury, there is presently no reliable treatment. The main factors driving brain damage from ischemic stroke are neuronal cell death resulting from oxidative stress, inflammation, and failure of the blood brain barrier. While under normal conditions, the blood brain barrier acts as a selectively permeable membrane allowing solutes and other substances to pass into the tissues of the central nervous system, ischemia and reperfusion alter the expression of tight junction proteins such as occludin, which leads to unmitigated perfusion and loss of homeostasis. Phoenixin-14 is a 14-amino acid neuropeptide that has been shown to play a role in regulating reproduction, blood sugar metabolism, pain, anxiety, and more recently, certain aspects of ischemic cardiac injury. In the present study, we found that phoenixin-14 confers protective effects against oxygen-glucose deprivation/reoxygenation (OGD/R) injury in bEnd.3 brain endothelial cells. Phoenixin-14 attenuated oxidative stress via downregulation of ROS and NOX1 and inhibited HMGB1 expression. Additionally, phoenixin-14 increased the expression of eNOS and NO, which play a protective role. Phoenixin-14 reduced endothelial monolayer permeability by increasing the expression of occludin. Finally, we found that the effects of phoenixin-14 on the expression of eNOS and occludin are dependent on the KLF2 transcriptional pathway, as evidenced by the results of our KLF2 knockdown experiment. Thus, phoenixin-14 may serve as a novel therapeutic agent for ischemic stroke.
    Keywords:  Blood brain barrier; KLF2; Occludin; Permeability; Phoenixin-14; Stroke
    DOI:  https://doi.org/10.1016/j.abb.2020.108275