138 results on '"Ushio-Fukai M"'
Search Results
2. Convergence of redox-sensitive and mitogen activated protein kinase signaling pathways in TNFalfa mediated MCP-1 induction in vascular smooth muscle cells
- Author
-
De Keulenaer, Gilles, Ushio-Fukai, M., QinQin, Y., Chung, A.B., Lyons, P.R., Ishizaka, N., Rengarajan, K., Taylor, W.R., Alexander, R.W., and Griendling, K.K.
- Published
- 2000
3. TNFalpha activates a p22phox-based NADH oxidase in vascular smooth muscle cells
- Author
-
De Keulenaer, Gilles, Ushio-Fukai, M., Ishizaka, N., Alexander, R.W., and Griendling, K.K.
- Published
- 1998
4. Effects of isoprenaline on cytosolic calcium concentrations and on tension in the porcine coronary artery.
- Author
-
Ushio-Fukai, M, primary, Abe, S, additional, Kobayashi, S, additional, Nishimura, J, additional, and Kanaide, H, additional
- Published
- 1993
- Full Text
- View/download PDF
5. Role of copper transport protein antioxidant 1 in angiotensin II-induced hypertension: a key regulator of extracellular superoxide dismutase.
- Author
-
Ozumi K, Sudhahar V, Kim HW, Chen GF, Kohno T, Finney L, Vogt S, McKinney RD, Ushio-Fukai M, Fukai T, Ozumi, Kiyoshi, Sudhahar, Varadarajan, Kim, Ha Won, Chen, Gin-Fu, Kohno, Takashi, Finney, Lydia, Vogt, Stefan, McKinney, Ronald D, Ushio-Fukai, Masuko, and Fukai, Tohru
- Abstract
Extracellular superoxide dismutase (SOD3) is a secretory copper enzyme involved in protecting angiotensin II (Ang II)-induced hypertension. We found previously that Ang II upregulates SOD3 expression and activity as a counterregulatory mechanism; however, underlying mechanisms are unclear. Antioxidant 1 (Atox1) is shown to act as a copper-dependent transcription factor, as well as a copper chaperone, for SOD3 in vitro, but its role in Ang II-induced hypertension in vivo is unknown. Here we show that Ang II infusion increases Atox1 expression, as well as SOD3 expression and activity, in aortas of wild-type mice, which are inhibited in mice lacking Atox1. Accordingly, Ang II increases vascular superoxide production, reduces endothelium-dependent vasodilation, and increases vasoconstriction in mesenteric arteries to a greater extent in Atox1(-/-) than in wild-type mice. This contributes to augmented hypertensive response to Ang II in Atox1(-/-) mice. In cultured vascular smooth muscle cells, Ang II promotes translocation of Atox1 to the nucleus, thereby increasing SOD3 transcription by binding to Atox1-responsive element in the SOD3 promoter. Furthermore, Ang II increases Atox1 binding to the copper exporter ATP7A, which obtains copper from Atox1, as well as translocation of ATP7A to plasma membranes, where it colocalizes with SOD3. As its consequence, Ang II decreases vascular copper levels, which is inhibited in Atox1(-/-) mice. In summary, Atox1 functions to prevent Ang II-induced endothelial dysfunction and hypercontraction in resistant vessels, as well as hypertension, in vivo by reducing extracellular superoxide levels via increasing vascular SOD3 expression and activity. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
6. Vascular signaling through G protein-coupled receptors: new concepts.
- Author
-
Ushio-Fukai M and Ushio-Fukai, Masuko
- Published
- 2009
- Full Text
- View/download PDF
7. Role of gp91phox (N0x2)-containing NAD(P)H oxidase in angiogenesis in response to hindlimb ischemia.
- Author
-
Tojo T, Ushio-Fukai M, Yamaoka-Tojo M, Ikeda S, Patrushev N, and Alexander RW
- Published
- 2005
8. The mechanism of the decrease in cytosolic Ca2+ concentrations induced by angiotensin II in the high K(+)-depolarized rabbit femoral artery.
- Author
-
Ushio-Fukai, Masuko, Yamamoto, Hiromichi, Nishimura, Junji, Hirano, Katsuya, Kanaide, Hideo, Ushio-Fukai, M, Yamamoto, H, Nishimura, J, Hirano, K, and Kanaide, H
- Published
- 2000
- Full Text
- View/download PDF
9. Role of NADH/NADPH oxidase-derived H2O2 in angiotensin II-induced vascular hypertrophy.
- Author
-
Zafari, A. Maziar, Ushio-Fukai, Masuko, Akers, Marjorie, Yin, Qiqin, Shah, Aalok, Harrison, David G., Taylor, W. Robert, Griendling, Kathy K., Zafari, A M, Ushio-Fukai, M, Akers, M, Yin, Q, Shah, A, Harrison, D G, Taylor, W R, and Griendling, K K
- Published
- 1998
10. Reactive oxygen species as mediators of angiotensin II signaling
- Author
-
Griendling, K. K. and Ushio-Fukai, M.
- Published
- 2000
- Full Text
- View/download PDF
11. Reactive oxygen species mediate the activation of Akt/protein kinase B by angiotensin II in vascular smooth muscle cells.
- Author
-
Ushio-Fukai, M, Alexander, R W, Akers, M, Yin, Q, Fujio, Y, Walsh, K, and Griendling, K K
- Abstract
Angiotensin II, a hypertrophic/anti-apoptotic hormone, utilizes reactive oxygen species (ROS) as growth-related signaling molecules in vascular smooth muscle cells (VSMCs). Recently, the cell survival protein kinase Akt/protein kinase B (PKB) was proposed to be involved in protein synthesis. Here we show that angiotensin II causes rapid phosphorylation of Akt/PKB (6- +/- 0.4-fold increase). Exogenous H(2)O(2) (50-200 microM) also stimulates Akt/PKB phosphorylation (maximal 8- +/- 0.2-fold increase), suggesting that Akt/PKB activation is redox-sensitive. Both angiotensin II and H(2)O(2) stimulation of Akt/PKB are abrogated by the phosphatidylinositol 3-kinase (PI3-K) inhibitors wortmannin and LY294002 (2(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one), suggesting that PI3-K is an upstream mediator of Akt/PKB activation in VSMCs. Furthermore, diphenylene iodonium, an inhibitor of flavin-containing oxidases, or overexpression of catalase to block angiotensin II-induced intracellular H(2)O(2) production significantly inhibits angiotensin II-induced Akt/PKB phosphorylation, indicating a role for ROS in agonist-induced Akt/PKB activation. In VSMCs infected with dominant-negative Akt/PKB, angiotensin II-stimulated [(3)H]leucine incorporation is attenuated. Thus, our studies indicate that Akt/PKB is part of the remarkable spectrum of angiotensin II signaling pathways and provide insight into the highly organized signaling mechanisms coordinated by ROS, which mediate the hypertrophic response to angiotensin II in VSMCs.
- Published
- 1999
12. p38 Mitogen-activated protein kinase is a critical component of the redox-sensitive signaling pathways activated by angiotensin II. Role in vascular smooth muscle cell hypertrophy.
- Author
-
Ushio-Fukai, M, Alexander, R W, Akers, M, and Griendling, K K
- Abstract
Angiotensin II induces an oxidant stress-dependent hypertrophy in cultured vascular smooth muscle cells. To investigate the growth-related molecular targets of H2O2, we examined the redox sensitivity of agonist-stimulated activation of the mitogen-activated protein kinase (MAPK) family. We show here that angiotensin II elicits a rapid increase in intracellular H2O2 and a rapid and robust phosphorylation of both p42/44MAPK (16-fold) and p38MAPK (15-fold). However, exogenous H2O2 activates only p38MAPK (14-fold), and diphenylene iodonium, an NADH/NADPH oxidase inhibitor, attenuates angiotensin II-stimulated phosphorylation of p38MAPK, but not p42/44MAPK. Furthermore, in cells stably transfected with human catalase, angiotensin II-induced intracellular H2O2 generation is almost completely blocked, resulting in inhibition of phosphorylation of p38MAPK, but not p42/44MAPK, and a subsequent partial decrease in angiotensin II-induced hypertrophy. Specific inhibition of either the p38MAPK pathway with SB203580 (4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H- imidaz ole) or the p42/44MAPK pathway with PD98059 (2-(2'-amino-3'-methoxyphenyl)oxanaphthalen-4-one) also partially, but significantly, attenuates angiotensin II-induced hypertrophy; however, simultaneous blockade of both pathways has an additive inhibitory effect, indicating that the hypertrophic response to angiotensin II requires parallel, independent activation of both MAPK pathways. These results provide the first evidence that p38MAPK is a critical component of the oxidant stress (H2O2)-sensitive signaling pathways activated by angiotensin II in vascular smooth muscle cells and indicate that it plays a crucial role in vascular hypertrophy.
- Published
- 1998
13. NADH/NADPH Oxidase and Vascular Function
- Author
-
Griendling, K. K. and Ushio-Fukai, M.
- Published
- 1997
- Full Text
- View/download PDF
14. Temporal dispersion of activation of phospholipase C-beta1 and -gamma isoforms by angiotensin II in vascular smooth muscle cells. Role of alphaq/11, alpha12, and beta gamma G protein subunits.
- Author
-
Ushio-Fukai, M, Griendling, K K, Akers, M, Lyons, P R, and Alexander, R W
- Abstract
Activation of phospholipase C (PLC) is one of the earliest events in angiotensin II (Ang II) type 1 (AT1) receptor (R)-mediated signal transduction in vascular smooth muscle cells (VSMCs). The coupling mechanisms of AT1 Rs to PLC, however, are controversial, because both tyrosine phosphorylation of PLC-gamma and G protein-dependent PLC-beta activation pathways have been reported. The expression of PLC-beta1, furthermore, has not been consistently demonstrated in VSMCs. Here we identified the PLC subtypes and subunits of heterotrimeric G proteins involved in AT1 R-PLC coupling using cultured rat VSMCs. Western analysis revealed the expression of PLC-beta1, -gamma1, and -delta1 in VSMCs. Ang II-stimulated inositol trisphosphate (IP3) formation measured at 15 s, which corresponds to the peak response, was significantly inhibited by electroporation of antibodies against PLC-beta1, but not by anti-PLC-gamma and -delta antibodies. Electroporation of anti-Galphaq/11 and -Galpha12 antibodies also showed significant inhibition of the Ang II-induced IP3 generation at 15 s, while anti-Galphai and Galpha13 antibodies were ineffective. Furthermore, in VSMCs electroporated with anti-Gbeta antibody and cells stably transfected with the plasmid encoding the Gbetagamma-binding region of the carboxyl terminus of beta-adrenergic receptor kinase1, the peak Ang II-stimulated PLC activity (at 15 s) was significantly inhibited. The tyrosine kinase inhibitor, genistein, had no effect on the peak response to Ang II stimulation, but significantly inhibited IP3 production after 30 s, a time period which temporally correlated with PLC-gamma tyrosine phosphorylation in response to Ang II. Moreover, electropor-ation of anti-PLC-gamma antibody markedly inhibited the IP3 production measured at 30 s, indicating that tyrosine phosphorylation of PLC-gamma contributes mainly to the later phase of PLC activation. Thus, these results suggest that: 1) AT1 receptors sequentially couple to PLC-beta1 via a heterotrimeric G protein and to PLC-gamma via a downstream tyrosine kinase; 2) the initial AT1 receptor-PLC-beta1 coupling is mediated by Galphaq/11beta gamma and Galpha12 beta gamma; 3) Gbeta gamma acts as a signal transducer for activation of PLC in VSMCs. The sequential coupling of AT1 receptors to PLC-beta1 and PLC-gamma, as well as dual coupling of AT1 receptors to distinct Galpha proteins, suggests a novel mechanism for a temporally controlled, highly organized and convergent Ang II-signaling network in VSMCs.
- Published
- 1998
15. Altered copper transport in oxidative stress-dependent brain endothelial barrier dysfunction associated with Alzheimer's disease.
- Author
-
Hossain MS, Das A, Rafiq AM, Deák F, Bagi Z, Outlaw R, Sudhahar V, Yamamoto M, Kaplan JH, Ushio-Fukai M, and Fukai T
- Abstract
Oxidative stress and blood-brain barrier (BBB) disruption due to brain endothelial barrier dysfunction contribute to Alzheimer's Disease (AD), which is characterized by beta-amyloid (Aβ) accumulation in senile plaques. Copper (Cu) is implicated in AD pathology and its levels are tightly controlled by several Cu transport proteins. However, their expression and role in AD, particularly in relation to brain endothelial barrier function remains unclear. In this study, we examined the expression of Cu transport proteins in the brains of AD mouse models as well as their involvement in Aβ42-induced brain endothelial barrier dysfunction. We found that the Cu uptake transporter CTR1 was upregulated, while the Cu exporter ATP7A was downregulated in the hippocampus of AD mouse models and in Aβ42-treated human brain microvascular endothelial cells (hBMECs). In the 5xFAD AD mouse model, Cu levels (assessed by ICP-MS) were elevated in the hippocampus. Moreover, in cultured hBMECs, Aβ42-induced reactive oxygen species (ROS) production, ROS-dependent loss in barrier function (measured by transendothelial electrical resistance), and tyrosine phosphorylation of CDH5 were all inhibited by either a membrane permeable Cu chelator or by knocking down CTR1 expression. These findings suggest that dysregulated expression of Cu transport proteins may lead to intracellular Cu accumulation in the AD brain, and that Aβ42 promotes ROS-dependent brain endothelial barrier dysfunction and CDH5 phosphorylation in a CTR1-Cu-dependent manner. Our study uncovers the critical role of Cu transport proteins in oxidative stress-related loss of BBB integrity in AD., Competing Interests: Declaration of competing interest The author (MUF) is an Editorial Board Member for Vascular Phar macology and was not involved in the editorial review or the decision to publish this article. All other authors (MSH, AD, AMR, FD, ZB, RO, VS, MY, JHK, TF) declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
16. Endothelial Drp1 Couples VEGF-induced Redox Signaling with Glycolysis Through Cysteine Oxidation to Drive Angiogenesis.
- Author
-
Nagarkoti S, Kim YM, Das A, Ash D, A Vitriol E, Read TA, Sudhahar V, Hossain MS, Yadav S, McMenamin M, Kelley S, Lucas R, Stepp D, Belin de Chantemele EJ, Caldwell RB, Fulton DJ, Fukai T, and Ushio-Fukai M
- Abstract
Angiogenesis plays a vital role for postnatal development and tissue repair following ischemia. Reactive oxygen species (ROS) generated by NADPH oxidases (NOXes) and mitochondria act as signaling molecules that promote angiogenesis in endothelial cells (ECs) which mainly relies on aerobic glycolysis for ATP production. However, the connections linking redox signaling with glycolysis are not well understood. The GTPase Drp1 is a member of the dynamin superfamily that moves from cytosol to mitochondria through posttranslational modifications to induce mitochondrial fission. The role of Drp1 in ROS-dependent VEGF signaling and angiogenesis in ECs has not been previously described. Here, we identify an unexpected function of endothelial Drp1 as a redox sensor, transmitting VEGF-induced H
2 O2 signals to enhance glycolysis and angiogenesis. Loss of Drp1 expression in ECs inhibited VEGF-induced angiogenic responses. Mechanistically, VEGF rapidly induced the NOX4-dependent sulfenylation (CysOH) of Drp1 on Cys644 , promoting disulfide bond formation with the metabolic kinase AMPK and subsequent sulfenylation of AMPK at Cys299 / 304 via the mitochondrial fission-mitoROS axis. This cysteine oxidation of AMPK, in turn, enhanced glycolysis and angiogenesis. In vivo , mice with EC-specific Drp1 deficiency or CRISPR/Cas9-engineered "redox-dead" (Cys to Ala) Drp1 knock-in mutations exhibited impaired retinal angiogenesis and post-ischemic neovascularization. Our findings uncover a novel role for endothelial Drp1 in linking VEGF-induced mitochondrial redox signaling to glycolysis through a cysteine oxidation-mediated Drp1-AMPK redox relay, driving both developmental and reparative angiogenesis.- Published
- 2024
- Full Text
- View/download PDF
17. Myeloid Drp1 Deficiency Limits Revascularization in Ischemic Muscles via Inflammatory Macrophage Polarization and Metabolic Reprograming.
- Author
-
Yadav S, Ganta V, Sudhahar V, Ash D, Nagarkoti S, Das A, McMenamin M, Kelley S, Fukai T, and Ushio-Fukai M
- Abstract
In the preclinical model of peripheral arterial disease (PAD), M2-like anti-inflammatory macrophage polarization and angiogenesis are required for revascularization. The regulation of cell metabolism and inflammation in macrophages is tightly linked to mitochondrial dynamics. Drp1, a mitochondrial fission protein, has shown context-dependent macrophage phenotypes with both pro- and anti-inflammatory characteristics. However, the role of macrophage Drp1 in reparative neovascularization remains unexplored. Here we show that Drp1 expression was significantly increased in F4/80+ macrophages within ischemic muscle at day 3 following hindlimb ischemia (HLI), an animal model of PAD. Myeloid-specific Drp1
-/- mice exhibited reduced limb perfusion recovery, angiogenesis and muscle regeneration after HLI. These effects were concomitant with enhancement of pro-inflammatory M1-like macrophages, p-NFkB, and TNFα levels, while showing reduction in anti-inflammatory M2-like macrophages and p-AMPK in ischemic muscle of myeloid Drp1-/- mice. In vitro, Drp1-/- macrophages under hypoxia serum starvation (HSS), an in vitro PAD model, demonstrated enhanced glycolysis via reducing p-AMPK as well as mitochondrial dysfunction and excessive mitochondrial ROS, resulting in increased M1-gene and reduced M2-gene expression. Conditioned media from HSS-treated Drp1-/- macrophages exhibited increased secretion of pro-inflammatory cytokines and suppressed angiogenic responses in cultured endothelial cells. Thus, Drp1 deficiency in macrophages under ischemia drives inflammatory metabolic reprogramming and macrophage polarization, thereby limiting revascularization in experimental PAD.- Published
- 2023
- Full Text
- View/download PDF
18. Type 1 Diabetes Impairs Endothelium-Dependent Relaxation Via Increasing Endothelial Cell Glycolysis Through Advanced Glycation End Products, PFKFB3, and Nox1-Mediated Mechanisms.
- Author
-
Atawia RT, Batori RK, Jordan CR, Kennard S, Antonova G, Bruder-Nascimento T, Mehta V, Saeed MI, Patel VS, Fukai T, Ushio-Fukai M, Huo Y, Fulton DJR, and Belin de Chantemèle EJ
- Subjects
- Animals, Mice, Pyruvaldehyde, Glycolysis, Endothelium, Endothelial Cells, Diabetes Mellitus, Type 1
- Abstract
Background: Type 1 diabetes (T1D) is a major cause of endothelial dysfunction. Although cellular bioenergetics has been identified as a new regulator of vascular function, whether glycolysis, the primary bioenergetic pathway in endothelial cells (EC), regulates vascular tone and contributes to impaired endothelium-dependent relaxation (EDR) in T1D remains unknown., Methods: Experiments were conducted in Akita mice with intact or selective deficiency in EC PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3), the main regulator of glycolysis. Seahorse analyzer and myography were employed to measure glycolysis and mitochondrial respiration, and EDR, respectively, in aortic explants. EC PFKFB3 (Ad-PFKFB3) and glycolysis (Ad-GlycoHi) were increased in situ via adenoviral transduction., Results: T1D increased EC glycolysis and elevated EC expression of PFKFB3 and NADPH oxidase Nox1 (NADPH oxidase homolog 1). Functionally, pharmacological and genetic inhibition of PFKFB3 restored EDR in T1D, while in situ aorta EC transduction with Ad-PFKFB3 or Ad-GlycoHi reproduced the impaired EDR associated with T1D. Nox1 inhibition restored EDR in aortic rings from Akita mice, as well as in Ad-PFKFB3-transduced aorta EC and lactate-treated wild-type aortas. T1D increased the expression of the advanced glycation end product precursor methylglyoxal in the aortas. Exposure of the aortas to methylglyoxal impaired EDR, which was prevented by PFKFB3 inhibition. T1D and exposure to methylglyoxal increased EC expression of HIF1α (hypoxia-inducible factor 1α), whose inhibition blunted methylglyoxal-mediated EC PFKFB3 upregulation., Conclusions: EC bioenergetics, namely glycolysis, is a new regulator of vasomotion and excess glycolysis, a novel mechanism of endothelial dysfunction in T1D. We introduce excess methylglyoxal, HIF1α, and PFKFB3 as major effectors in T1D-mediated increased EC glycolysis., Competing Interests: Disclosures None.
- Published
- 2023
- Full Text
- View/download PDF
19. Pentose Pathway Activation Is Superior to Increased Glycolysis for Therapeutic Angiogenesis in Peripheral Arterial Disease.
- Author
-
Zaied AA, Ushio-Fukai M, Fukai T, Kovacs-Kasa A, Alhusban S, Sudhahar V, Ganta VC, and Annex BH
- Subjects
- Mice, Animals, Endothelial Cells metabolism, Vascular Endothelial Growth Factor A metabolism, Reactive Oxygen Species metabolism, Hypoxia metabolism, Glycolysis physiology, Ischemia genetics, Peripheral Arterial Disease metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
Background In endothelial cells (ECs), glycolysis, regulated by PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase, isoform-3), is the major metabolic pathway for ATP generation. In preclinical peripheral artery disease models, VEGF
165 a (vascular endothelial growth factor165 a) and microRNA-93 both promote angiogenesis. Methods and Results Mice following hind-limb ischemia (HLI) and ECs with, and without, hypoxia and serum starvation were examined with, and without, microRNA-93 and VEGF165 a. Post-HLI perfusion recovery was monitored. EC metabolism was studied using seahorse assay, and the expression and activity of major metabolism genes were assessed. Reactive oxygen species levels and EC permeability were evaluated. C57Bl/6J mice generated a robust angiogenic response to HLI, with ECs from ischemic versus nonischemic muscle demonstrating no increase in glycolysis. Balb/CJ mice generated a poor angiogenic response post-HLI; ischemic versus nonischemic ECs demonstrated significant increase in glycolysis. MicroRNA-93-treated Balb/CJ mice post-HLI showed better perfusion recovery, with ischemic versus nonischemic ECs showing no increase in glycolysis. VEGF165 a-treated Balb/CJ mice post-HLI showed no improvement in perfusion recovery with ischemic versus nonischemic ECs showing significant increase in glycolysis. ECs under hypoxia and serum starvation upregulated PFKFB3. In ECs under hypoxia and serum starvation, VEGF165 a versus control significantly upregulated PFKFB3 and glycolysis, whereas miR-93 versus control demonstrated no increase in PFKFB3 or glycolysis. MicroRNA-93 versus VEGF165 a upregulated glucose-6-phosphate dehydrogenase expression and activity, activating the pentose phosphate pathway. MicroRNA-93 versus control increased reduced nicotinamide adenine dinucleotide phosphate and virtually eliminated the increase in reactive oxygen species. In ECs under hypoxia and serum starvation, VEGF165 a significantly increased and miR-93 decreased EC permeability. Conclusions In peripheral artery disease, activation of the pentose phosphate pathway to promote angiogenesis may offer potential therapeutic advantages.- Published
- 2023
- Full Text
- View/download PDF
20. Protein disulfide isomerase A1 as a novel redox sensor in VEGFR2 signaling and angiogenesis.
- Author
-
Nagarkoti S, Kim YM, Ash D, Das A, Vitriol E, Read TA, Youn SW, Sudhahar V, McMenamin M, Hou Y, Boatwright H, Caldwell R, Essex DW, Cho J, Fukai T, and Ushio-Fukai M
- Subjects
- Mice, Humans, Animals, Reactive Oxygen Species metabolism, Vascular Endothelial Growth Factor A metabolism, Hydrogen Peroxide metabolism, Neovascularization, Physiologic, Oxidation-Reduction, Vascular Endothelial Growth Factor Receptor-2 genetics, Vascular Endothelial Growth Factor Receptor-2 metabolism, Ischemia metabolism, Endothelial Cells metabolism, Protein Disulfide-Isomerases genetics, Protein Disulfide-Isomerases metabolism
- Abstract
VEGFR2 signaling in endothelial cells (ECs) is regulated by reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria, which plays an important role in postnatal angiogenesis. However, it remains unclear how highly diffusible ROS signal enhances VEGFR2 signaling and reparative angiogenesis. Protein disulfide isomerase A1 (PDIA1) functions as an oxidoreductase depending on the redox environment. We hypothesized that PDIA1 functions as a redox sensor to enhance angiogenesis. Here we showed that PDIA1 co-immunoprecipitated with VEGFR2 or colocalized with either VEGFR2 or an early endosome marker Rab5 at the perinuclear region upon stimulation of human ECs with VEGF. PDIA1 silencing significantly reduced VEGF-induced EC migration, proliferation and spheroid sprouting via inhibiting VEGFR2 signaling. Mechanistically, VEGF stimulation rapidly increased Cys-OH formation of PDIA1 via the NOX4-mitochondrial ROS axis. Overexpression of "redox-dead" mutant PDIA1 with replacement of the active four Cys residues with Ser significantly inhibited VEGF-induced PDIA1-CysOH formation and angiogenic responses via reducing VEGFR2 phosphorylation. Pdia1
+/- mice showed impaired angiogenesis in developmental retina and Matrigel plug models as well as ex vivo aortic ring sprouting model. Study using hindlimb ischemia model revealed that PDIA1 expression was markedly increased in angiogenic ECs of ischemic muscles, and that ischemia-induced limb perfusion recovery and neovascularization were impaired in EC-specific Pdia1 conditional knockout mice. These results suggest that PDIA1 can sense VEGF-induced H2 O2 signal via CysOH formation to promote VEGFR2 signaling and angiogenesis in ECs, thereby enhancing postnatal angiogenesis. The oxidized PDIA1 is a potential therapeutic target for treatment of ischemic vascular diseases., (© 2022. The Author(s), under exclusive licence to Springer Nature B.V.)- Published
- 2023
- Full Text
- View/download PDF
21. Whole-Transcriptome Sequencing Analyses of Nuclear Antixoxidant-1 in Endothelial Cells: Role in Inflammation and Atherosclerosis.
- Author
-
Sudhahar V, Shi Y, Kaplan JH, Ushio-Fukai M, and Fukai T
- Subjects
- Animals, Copper Transport Proteins, Cytokines metabolism, Endothelial Cells metabolism, Humans, Inflammation genetics, Mice, Mice, Knockout, ApoE, Molecular Chaperones metabolism, Reactive Oxygen Species metabolism, Transcriptome, Atherosclerosis genetics, Copper metabolism
- Abstract
Inflammation, oxidative stress, and copper (Cu) play an important role in cardiovascular disease, including atherosclerosis. We previously reported that cytosolic Cu chaperone antioxidant-1 (Atox1) translocates to the nucleus in response to inflammatory cytokines or exogenous Cu and that Atox1 is localized at the nucleus in the endothelium of inflamed atherosclerotic aorta. However, the roles of nuclear Atox1 and their function are poorly understood. Here we showed that Atox1 deficiency in ApoE
-/- mice with a Western diet exhibited a significant reduction of atherosclerotic lesion formation. In vitro, adenovirus-mediated overexpression of nuclear-targeted Atox1 (Ad-Atox1-NLS) in cultured human endothelial cells (ECs) increased monocyte adhesion and reactive oxygen species (ROS) production compared to control cells (Ad-null). To address the underlying mechanisms, we performed genome-wide mapping of Atox1-regulated targets in ECs, using an unbiased systemic approach integrating sequencing data. Combination of ChIP-Seq and RNA-Seq analyses in ECs transfected with Ad-Atox1-NLS or Ad-null identified 1387 differentially expressed genes (DEG). Motif enrichment assay and KEGG pathway enrichment analysis revealed that 248 differentially expressed genes, including inflammatory and angiogenic genes, were regulated by Atox1-NLS, which was then confirmed by real-time qPCR. Among these genes, functional analysis of inflammatory responses identified CD137, CSF1, and IL5RA as new nuclear Atox1-targeted inflammatory genes, while CD137 is also a key regulator of Atox1-NLS-induced ROS production. These findings uncover new nuclear Atox1 downstream targets involved in inflammation and ROS production and provide insights into the nuclear Atox1 as a potential therapeutic target for the treatment of inflammatory diseases such as atherosclerosis.- Published
- 2022
- Full Text
- View/download PDF
22. Editorial: Mitochondria, metabolism and cardiovascular diseases.
- Author
-
Koga JI, Sun X, and Ushio-Fukai M
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
23. Embracing Diversity, Equity, and Inclusion in the Scientific Community-Viewpoints of the Diversity, Equity, and Inclusion Committee of the North American Vascular Biology Organization.
- Author
-
Garelnabi M, Cowdin M, Fang Y, Shrestha B, Ushio-Fukai M, Aikawa E, Graham G, Molema G, Yanagisawa H, and Aikawa M
- Abstract
Recent increased visibility on racial issues in the United States elicited public outcry and a collective call for action. The social justice movement has facilitated energetic discussions about race, sexual orientation, and various issues of diversity, equity, and inclusion. This article discusses issues faced by people of color that we as scientists can address, as well as challenges faced by women and internationally trained scientists in the scientific community that need immediate attention. Moreover, we highlight various ways to resolve such issues at both institutional and individual levels. Silence and incremental solutions are no longer acceptable to achieving lasting social justice and ensure prosperous societies that work for all., Competing Interests: GG was employed by Healthcare and Public Health at Google/YouTube. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Garelnabi, Cowdin, Fang, Shrestha, Ushio-Fukai, Aikawa, Graham, Molema, Yanagisawa and Aikawa.)
- Published
- 2022
- Full Text
- View/download PDF
24. Role of prostaglandin D2 receptors in the pathogenesis of abdominal aortic aneurysm formation.
- Author
-
Weintraub NL, Blomkalns AL, Ogbi M, Horimatsu T, Benson TW, Huo Y, Fulton DJ, Agarwal G, Lee R, Winkler MA, Young L, Fujise K, Guha A, Fukai T, Ushio-Fukai M, Long X, Annex BH, and Kim HW
- Subjects
- Angiotensin II pharmacology, Animals, Aortic Aneurysm, Abdominal prevention & control, Male, Mice, Receptors, Immunologic antagonists & inhibitors, Receptors, Prostaglandin antagonists & inhibitors, Aortic Aneurysm, Abdominal etiology, Receptors, Immunologic physiology, Receptors, Prostaglandin physiology
- Abstract
Prostaglandin D2 (PGD2) released from immune cells or other cell types activates its receptors, D prostanoid receptor (DP)1 and 2 (DP1 and DP2), to promote inflammatory responses in allergic and lung diseases. Prostaglandin-mediated inflammation may also contribute to vascular diseases such as abdominal aortic aneurysm (AAA). However, the role of DP receptors in the pathogenesis of AAA has not been systematically investigated. In the present study, DP1-deficient mice and pharmacological inhibitors of either DP1 or DP2 were tested in two distinct mouse models of AAA formation: angiotensin II (AngII) infusion and calcium chloride (CaCl2) application. DP1-deficient mice [both heterozygous (DP1+/-) and homozygous (DP1-/-)] were protected against CaCl2-induced AAA formation, in conjunction with decreased matrix metallopeptidase (MMP) activity and adventitial inflammatory cell infiltration. In the AngII infusion model, DP1+/- mice, but not DP1-/- mice, exhibited reduced AAA formation. Interestingly, compensatory up-regulation of the DP2 receptor was detected in DP1-/- mice in response to AngII infusion, suggesting a potential role for DP2 receptors in AAA. Treatment with selective antagonists of DP1 (laropiprant) or DP2 (fevipiprant) protected against AAA formation, in conjunction with reduced elastin degradation and aortic inflammatory responses. In conclusion, PGD2 signaling contributes to AAA formation in mice, suggesting that antagonists of DP receptors, which have been extensively tested in allergic and lung diseases, may be promising candidates to ameliorate AAA., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
25. Exercise improves angiogenic function of circulating exosomes in type 2 diabetes: Role of exosomal SOD3.
- Author
-
Abdelsaid K, Sudhahar V, Harris RA, Das A, Youn SW, Liu Y, McMenamin M, Hou Y, Fulton D, Hamrick MW, Tang Y, Fukai T, and Ushio-Fukai M
- Subjects
- Animals, Cells, Cultured, Copper-Transporting ATPases blood, Copper-Transporting ATPases metabolism, Diabetes Mellitus, Type 2 physiopathology, Endothelium, Vascular metabolism, Endothelium, Vascular physiology, Exercise, Female, Humans, Male, Mice, Mice, Inbred C57BL, Middle Aged, Physical Conditioning, Animal methods, Rats, Superoxide Dismutase blood, Diabetes Mellitus, Type 2 metabolism, Exosomes metabolism, Neovascularization, Physiologic, Running, Superoxide Dismutase metabolism
- Abstract
Exosomes, key mediators of cell-cell communication, derived from type 2 diabetes mellitus (T2DM) exhibit detrimental effects. Exercise improves endothelial function in part via the secretion of exosomes into circulation. Extracellular superoxide dismutase (SOD3) is a major secretory copper (Cu) antioxidant enzyme that catalyzes the dismutation of O
2 •- to H2 O2 whose activity requires the Cu transporter ATP7A. However, the role of SOD3 in exercise-induced angiogenic effects of circulating plasma exosomes on endothelial cells (ECs) in T2DM remains unknown. Here, we show that both SOD3 and ATP7A proteins were present in plasma exosomes in mice, which was significantly increased after two weeks of volunteer wheel exercise. A single bout of exercise in humans also showed a significant increase in SOD3 and ATP7A protein expression in plasma exosomes. Plasma exosomes from T2DM mice significantly reduced angiogenic responses in human ECs or mouse skin wound healing models, which was associated with a decrease in ATP7A, but not SOD3 expression in exosomes. Exercise training in T2DM mice restored the angiogenic effects of T2DM exosomes in ECs by increasing ATP7A in exosomes, which was not observed in exercised T2DM/SOD3-/- mice. Furthermore, exosomes overexpressing SOD3 significantly enhanced angiogenesis in ECs by increasing local H2 O2 levels in a heparin-binding domain-dependent manner as well as restored defective wound healing and angiogenesis in T2DM or SOD3-/- mice. In conclusion, exercise improves the angiogenic potential of circulating exosomes in T2DM in a SOD3-dependent manner. Exosomal SOD3 may provide an exercise mimetic therapy that supports neovascularization and wound repair in cardiometabolic disease., (© 2022 Federation of American Societies for Experimental Biology.)- Published
- 2022
- Full Text
- View/download PDF
26. Dichotomous Role of Tumor Necrosis Factor in Pulmonary Barrier Function and Alveolar Fluid Clearance.
- Author
-
Lucas R, Hadizamani Y, Enkhbaatar P, Csanyi G, Caldwell RW, Hundsberger H, Sridhar S, Lever AA, Hudel M, Ash D, Ushio-Fukai M, Fukai T, Chakraborty T, Verin A, Eaton DC, Romero M, and Hamacher J
- Abstract
Alveolar-capillary leak is a hallmark of the acute respiratory distress syndrome (ARDS), a potentially lethal complication of severe sepsis, trauma and pneumonia, including COVID-19. Apart from barrier dysfunction, ARDS is characterized by hyper-inflammation and impaired alveolar fluid clearance (AFC), which foster the development of pulmonary permeability edema and hamper gas exchange. Tumor Necrosis Factor (TNF) is an evolutionarily conserved pleiotropic cytokine, involved in host immune defense against pathogens and cancer. TNF exists in both membrane-bound and soluble form and its mainly -but not exclusively- pro-inflammatory and cytolytic actions are mediated by partially overlapping TNFR1 and TNFR2 binding sites situated at the interface between neighboring subunits in the homo-trimer. Whereas TNFR1 signaling can mediate hyper-inflammation and impaired barrier function and AFC in the lungs, ligand stimulation of TNFR2 can protect from ventilation-induced lung injury. Spatially distinct from the TNFR binding sites, TNF harbors within its structure a lectin-like domain that rather protects lung function in ARDS. The lectin-like domain of TNF -mimicked by the 17 residue TIP peptide- represents a physiological mediator of alveolar-capillary barrier protection. and increases AFC in both hydrostatic and permeability pulmonary edema animal models. The TIP peptide directly activates the epithelial sodium channel (ENaC) -a key mediator of fluid and blood pressure control- upon binding to its α subunit, which is also a part of the non-selective cation channel (NSC). Activity of the lectin-like domain of TNF is preserved in complexes between TNF and its soluble TNFRs and can be physiologically relevant in pneumonia. Antibody- and soluble TNFR-based therapeutic strategies show considerable success in diseases such as rheumatoid arthritis, psoriasis and inflammatory bowel disease, but their chronic use can increase susceptibility to infection. Since the lectin-like domain of TNF does not interfere with TNF's anti-bacterial actions, while exerting protective actions in the alveolar-capillary compartments, it is currently evaluated in clinical trials in ARDS and COVID-19. A more comprehensive knowledge of the precise role of the TNFR binding sites versus the lectin-like domain of TNF in lung injury, tissue hypoxia, repair and remodeling may foster the development of novel therapeutics for ARDS., Competing Interests: RL is inventor of several patents relating to the use of the TNF-derived TIP peptide in pulmonary edema reabsorption. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Lucas, Hadizamani, Enkhbaatar, Csanyi, Caldwell, Hundsberger, Sridhar, Lever, Hudel, Ash, Ushio-Fukai, Fukai, Chakraborty, Verin, Eaton, Romero and Hamacher.)
- Published
- 2022
- Full Text
- View/download PDF
27. Cysteine oxidation of copper transporter CTR1 drives VEGFR2 signalling and angiogenesis.
- Author
-
Das A, Ash D, Fouda AY, Sudhahar V, Kim YM, Hou Y, Hudson FZ, Stansfield BK, Caldwell RB, McMenamin M, Littlejohn R, Su H, Regan MR, Merrill BJ, Poole LB, Kaplan JH, Fukai T, and Ushio-Fukai M
- Subjects
- Animals, Cattle, Cell Line, Copper Transporter 1 genetics, Cysteine metabolism, Female, HEK293 Cells, Human Umbilical Vein Endothelial Cells, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Oxidation-Reduction, Signal Transduction physiology, Copper metabolism, Copper Transporter 1 metabolism, Neovascularization, Physiologic physiology, Reactive Oxygen Species metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Vascular endothelial growth factor receptor type 2 (VEGFR2, also known as KDR and FLK1) signalling in endothelial cells (ECs) is essential for developmental and reparative angiogenesis. Reactive oxygen species and copper (Cu) are also involved in these processes. However, their inter-relationship is poorly understood. Evidence of the role of the endothelial Cu importer CTR1 (also known as SLC31A1) in VEGFR2 signalling and angiogenesis in vivo is lacking. Here, we show that CTR1 functions as a redox sensor to promote angiogenesis in ECs. CTR1-depleted ECs showed reduced VEGF-induced VEGFR2 signalling and angiogenic responses. Mechanistically, CTR1 was rapidly sulfenylated at Cys189 at its cytosolic C terminus after stimulation with VEGF, which induced CTR1-VEGFR2 disulfide bond formation and their co-internalization to early endosomes, driving sustained VEGFR2 signalling. In vivo, EC-specific Ctr1-deficient mice or CRISPR-Cas9-generated redox-dead Ctr1(C187A)-knockin mutant mice had impaired developmental and reparative angiogenesis. Thus, oxidation of CTR1 at Cys189 promotes VEGFR2 internalization and signalling to enhance angiogenesis. Our study uncovers an important mechanism for sensing reactive oxygen species through CTR1 to drive neovascularization., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
28. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease.
- Author
-
Ushio-Fukai M, Ash D, Nagarkoti S, Belin de Chantemèle EJ, Fulton DJR, and Fukai T
- Subjects
- Homeostasis, Humans, Metabolic Networks and Pathways, NADP metabolism, Signal Transduction, Cardiovascular Diseases metabolism, Reactive Nitrogen Species metabolism, Reactive Oxygen Species metabolism
- Abstract
Reactive oxygen species (ROS; e.g ., superoxide [O
2 •- ] and hydrogen peroxide [H2 O2 ]) and reactive nitrogen species (RNS; e.g ., nitric oxide [NO• ]) at the physiological level function as signaling molecules that mediate many biological responses, including cell proliferation, migration, differentiation, and gene expression. By contrast, excess ROS/RNS, a consequence of dysregulated redox homeostasis, is a hallmark of cardiovascular disease. Accumulating evidence suggests that both ROS and RNS regulate various metabolic pathways and enzymes. Recent studies indicate that cells have mechanisms that fine-tune ROS/RNS levels by tight regulation of metabolic pathways, such as glycolysis and oxidative phosphorylation. The ROS/RNS-mediated inhibition of glycolytic pathways promotes metabolic reprogramming away from glycolytic flux toward the oxidative pentose phosphate pathway to generate nicotinamide adenine dinucleotide phosphate (NADPH) for antioxidant defense. This review summarizes our current knowledge of the mechanisms by which ROS/RNS regulate metabolic enzymes and cellular metabolism and how cellular metabolism influences redox homeostasis and the pathogenesis of disease. A full understanding of these mechanisms will be important for the development of new therapeutic strategies to treat diseases associated with dysregulated redox homeostasis and metabolism. Antioxid. Redox Signal . 34, 1319-1354.- Published
- 2021
- Full Text
- View/download PDF
29. The P-type ATPase transporter ATP7A promotes angiogenesis by limiting autophagic degradation of VEGFR2.
- Author
-
Ash D, Sudhahar V, Youn SW, Okur MN, Das A, O'Bryan JP, McMenamin M, Hou Y, Kaplan JH, Fukai T, and Ushio-Fukai M
- Subjects
- Animals, Blood Vessels drug effects, Blood Vessels physiology, COS Cells, Cells, Cultured, Chlorocebus aethiops, Copper-Transporting ATPases metabolism, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelial Cells physiology, Humans, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Microtubule-Associated Proteins metabolism, P-type ATPases metabolism, RNA Interference, Signal Transduction genetics, Vascular Endothelial Growth Factor A pharmacology, Vascular Endothelial Growth Factor Receptor-2 metabolism, Mice, Autophagy genetics, Blood Vessels metabolism, Copper-Transporting ATPases genetics, P-type ATPases genetics, Vascular Endothelial Growth Factor Receptor-2 genetics
- Abstract
VEGFR2 (KDR/Flk1) signaling in endothelial cells (ECs) plays a central role in angiogenesis. The P-type ATPase transporter ATP7A regulates copper homeostasis, and its role in VEGFR2 signaling and angiogenesis is entirely unknown. Here, we describe the unexpected crosstalk between the Copper transporter ATP7A, autophagy, and VEGFR2 degradation. The functional significance of this Copper transporter was demonstrated by the finding that inducible EC-specific ATP7A deficient mice or ATP7A-dysfunctional ATP7Amut mice showed impaired post-ischemic neovascularization. In ECs, loss of ATP7A inhibited VEGF-induced VEGFR2 signaling and angiogenic responses, in part by promoting ligand-induced VEGFR2 protein degradation. Mechanistically, VEGF stimulated ATP7A translocation from the trans-Golgi network to the plasma membrane where it bound to VEGFR2, which prevented autophagy-mediated lysosomal VEGFR2 degradation by inhibiting autophagic cargo/adapter p62/SQSTM1 binding to ubiquitinated VEGFR2. Enhanced autophagy flux due to ATP7A dysfunction in vivo was confirmed by autophagy reporter CAG-ATP7Amut -RFP-EGFP-LC3 transgenic mice. In summary, our study uncovers a novel function of ATP7A to limit autophagy-mediated degradation of VEGFR2, thereby promoting VEGFR2 signaling and angiogenesis, which restores perfusion recovery and neovascularization. Thus, endothelial ATP7A is identified as a potential therapeutic target for treatment of ischemic cardiovascular diseases.
- Published
- 2021
- Full Text
- View/download PDF
30. Shear and Integrin Outside-In Signaling Activate NADPH-Oxidase 2 to Promote Platelet Activation.
- Author
-
Xu Z, Liang Y, Delaney MK, Zhang Y, Kim K, Li J, Bai Y, Cho J, Ushio-Fukai M, Cheng N, and Du X
- Subjects
- Animals, Enzyme Activation, Female, GTP-Binding Protein alpha Subunits, G12-G13 genetics, GTP-Binding Protein alpha Subunits, G12-G13 metabolism, Integrin beta3 genetics, Male, Mice, Inbred C57BL, Mice, Knockout, NADPH Oxidase 2 genetics, NADPH Oxidases metabolism, Phosphatidylinositol 3-Kinase metabolism, Phosphorylation, Platelet Factor 4 genetics, Platelet Factor 4 metabolism, Platelet Glycoprotein GPIIb-IIIa Complex genetics, Proto-Oncogene Proteins c-akt metabolism, Stress, Mechanical, Syk Kinase metabolism, Mice, Blood Platelets enzymology, Hydrogen Peroxide metabolism, Integrin beta3 metabolism, Mechanotransduction, Cellular, NADPH Oxidase 2 metabolism, Platelet Activation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
31. Caveolin-1 stabilizes ATP7A, a copper transporter for extracellular SOD, in vascular tissue to maintain endothelial function.
- Author
-
Sudhahar V, Okur MN, O'Bryan JP, Minshall RD, Fulton D, Ushio-Fukai M, and Fukai T
- Subjects
- Animals, Aorta cytology, Aorta metabolism, Caveolin 1 deficiency, Copper pharmacology, Copper Transport Proteins genetics, Copper Transport Proteins metabolism, Copper-Transporting ATPases metabolism, Endothelial Cells cytology, Endothelial Cells drug effects, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Gene Expression Regulation, Male, Mesenteric Arteries cytology, Mesenteric Arteries metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Molecular Chaperones genetics, Molecular Chaperones metabolism, Oxidative Stress, Primary Cell Culture, Proteasome Endopeptidase Complex metabolism, Proteolysis, Signal Transduction, Superoxide Dismutase metabolism, Superoxide Dismutase-1 metabolism, Ubiquitination drug effects, Vasodilation drug effects, Caveolin 1 genetics, Copper-Transporting ATPases genetics, Endothelial Cells metabolism, Superoxide Dismutase genetics, Superoxide Dismutase-1 genetics
- Abstract
Caveolin-1 (Cav-1) is a scaffolding protein and a major component of caveolae/lipid rafts. Previous reports have shown that endothelial dysfunction in Cav-1-deficient (Cav-1
-/- ) mice is mediated by elevated oxidative stress through endothelial nitric oxide synthase (eNOS) uncoupling and increased NADPH oxidase. Oxidant stress is the net balance of oxidant generation and scavenging, and the role of Cav-1 as a regulator of antioxidant enzymes in vascular tissue is poorly understood. Extracellular SOD (SOD3) is a copper (Cu)-containing enzyme that is secreted from vascular smooth muscle cells/fibroblasts and subsequently binds to the endothelial cells surface, where it scavenges extracellular [Formula: see text] and preserves endothelial function. SOD3 activity is dependent on Cu, supplied by the Cu transporter ATP7A, but whether Cav-1 regulates the ATP7A-SOD3 axis and its role in oxidative stress-mediated vascular dysfunction has not been studied. Here we show that the activity of SOD3, but not SOD1, was significantly decreased in Cav-1-/- vessels, which was rescued by re-expression of Cav-1 or Cu supplementation. Loss of Cav-1 reduced ATP7A protein, but not mRNA, and this was mediated by ubiquitination of ATP7A and proteasomal degradation. ATP7A bound to Cav-1 and was colocalized with SOD3 in caveolae/lipid rafts or perinucleus in vascular tissues or cells. Impaired endothelium-dependent vasorelaxation in Cav-1-/- mice was rescued by gene transfer of SOD3 or by ATP7A-overexpressing transgenic mice. These data reveal an unexpected role of Cav-1 in stabilizing ATP7A protein expression by preventing its ubiquitination and proteasomal degradation, thereby increasing SOD3 activity, which in turn protects against vascular oxidative stress-mediated endothelial dysfunction.- Published
- 2020
- Full Text
- View/download PDF
32. Cross-Talk between NADPH Oxidase and Mitochondria: Role in ROS Signaling and Angiogenesis.
- Author
-
Fukai T and Ushio-Fukai M
- Subjects
- Animals, Endothelial Cells cytology, Endothelial Cells pathology, Endothelium, Vascular cytology, Endothelium, Vascular pathology, Humans, Neovascularization, Pathologic, Neovascularization, Physiologic, Oxidation-Reduction, Oxidative Stress, Endothelial Cells metabolism, Endothelium, Vascular metabolism, Mitochondria metabolism, NADPH Oxidases metabolism, Reactive Oxygen Species metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Angiogenesis, a new vessel formation from the pre-existing ones, is essential for embryonic development, wound repair and treatment of ischemic heart and limb diseases. However, dysregulated angiogenesis contributes to various pathologies such as diabetic retinopathy, atherosclerosis and cancer. Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) as well as mitochondria play an important role in promoting the angiogenic switch from quiescent endothelial cells (ECs). However, how highly diffusible ROS produced from different sources and location can communicate with each other to regulate angiogenesis remains unclear. To detect a localized ROS signal in distinct subcellular compartments in real time in situ, compartment-specific genetically encoded redox-sensitive fluorescence biosensors have been developed. Recently, the intercellular communication, "cross-talk", between ROS derived from NOX and mitochondria, termed "ROS-induced ROS release", has been proposed as a mechanism for ROS amplification at distinct subcellular compartments, which are essential for activation of redox signaling. This "ROS-induced ROS release" may represent a feed-forward mechanism of localized ROS production to maintain sustained signaling, which can be targeted under pathological conditions with oxidative stress or enhanced to promote therapeutic angiogenesis. In this review, we summarize the recent knowledge regarding the role of the cross-talk between NOX and mitochondria organizing the sustained ROS signaling involved in VEGF signaling, neovascularization and tissue repair.
- Published
- 2020
- Full Text
- View/download PDF
33. IQ motif-containing GTPase-activating protein 1 is essential for the optimal maintenance of lung ILC2s.
- Author
-
Tayama S, Okuyama Y, Phung HT, Asao A, Kobayashi S, Musha T, Machiyama T, Sakurai T, Zhang C, Ushio-Fukai M, Kawabe T, So T, and Ishii N
- Subjects
- Animals, Homeostasis immunology, Immunity, Innate immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, ras GTPase-Activating Proteins deficiency, Lung immunology, Lymphocytes immunology, ras GTPase-Activating Proteins immunology
- Abstract
Group 2 innate lymphoid cells (ILC2s) play critical roles in type 2 immunity and are crucial for pathogenesis of various types of inflammatory disease. IQ motif-containing GTPase-activating protein 1 (IQGAP1) is a ubiquitously expressed scaffold protein that is involved in multiple cellular functions such as cell survival and trafficking. While the roles for IQGAP1 in T and B lymphocytes have been uncovered, the physiological significance of IQGAP1 in innate lymphocytes remains to be elucidated. In the current study, we demonstrate that using bone marrow chimeras, the deficiency of IQGAP1 caused an impaired survival of lung ILC2s in a cell-intrinsic manner and that Iqgap1-/- mice displayed decreased accumulation of ILC2s after administration of papain and thereby reduced the pathology of the disease. Moreover, Iqgap1-/- ILC2s showed a significantly enhanced apoptosis as compared to wild-type ILC2s under both steady-state and inflammatory conditions. Together these results identify for the first time that IQGAP1 is essential for homeostasis of ILC2s in the lung., (© The Japanese Society for Immunology. 2019. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
34. Nuclear translocation of Atox1 potentiates activin A-induced cell migration and colony formation in colon cancer.
- Author
-
Jana A, Das A, Krett NL, Guzman G, Thomas A, Mancinelli G, Bauer J, Ushio-Fukai M, Fukai T, and Jung B
- Subjects
- Cell Line, Tumor, Cell Proliferation, Humans, Activins metabolism, Carcinoma metabolism, Carcinoma pathology, Cell Movement, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Copper Transport Proteins physiology, Molecular Chaperones physiology
- Abstract
Background: Colorectal cancer remains a deadly cancer due to metastatic disease. To understand the molecular mechanisms of metastasis in colon cancer, we investigated whether the copper chaperone antioxidant-1 (Atox1) protein plays a role in this process. Recent findings indicate that Atox1 protein has transcription factor activities and plays a vital role in cell proliferation in cancer cells. However, the role of Atox1 in metastasis has not been examined., Methods: Atox1 expression was determined by immunofluorescence in a tissue microarray generated from a spectrum of CRC patients. Subcellular fractionation of colon cancer cell lines SW480 and SW620 cells was used to examine the cellular location of Atox1 in the face of activin A, a cytokine that stimulates colon cancer metastasis. Atox1 expression was genetically manipulated and cellular migration measured through trans-well assay and proliferation measured by colony formation assays., Results: Here we demonstrate that in patients with metastatic colon cancer, there is a significant increase in the expression of nuclear Atox1. Interestingly, the metastatic CRC cell line SW620 has increased nuclear localization of Atox1 compared to its related non-metastatic cell line SW480. Further, inhibition of endogenous Atox1 by siRNA in SW620 decreased colony formation and reactive oxygen species generation via decreased expression of Atox1 targets cyclin D1 and NADPH oxidase subunit p47 phox, respectively. Additionally, overexpression of nuclear-targeted but not copper binding domain-mutated Atox1 in SW480 cells increased colony formation and cell migration that was further augmented by activin A stimulation, a known enhancer of colon cancer metastasis., Conclusions: Our findings suggest that nuclear Atox1 might be a new therapeutic target as well as a new biomarker for metastatic colorectal cancer., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
35. IQGAP1 restrains T-cell cosignaling mediated by OX40.
- Author
-
Okuyama Y, Nagashima H, Ushio-Fukai M, Croft M, Ishii N, and So T
- Subjects
- Animals, CD4-Positive T-Lymphocytes metabolism, Cytokines metabolism, Encephalomyelitis, Autoimmune, Experimental metabolism, Encephalomyelitis, Autoimmune, Experimental pathology, Inflammation metabolism, Inflammation pathology, Inflammation Mediators metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Receptors, OX40 genetics, Signal Transduction, CD4-Positive T-Lymphocytes immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Immunologic Memory immunology, Inflammation immunology, Lymphocyte Activation immunology, Receptors, OX40 metabolism, ras GTPase-Activating Proteins physiology
- Abstract
A costimulatory signal from the tumor necrosis factor receptor (TNFR) family molecule OX40 (CD134), which is induced on activated T cells, is important for T-cell immunity. Aberrant OX40 cosignaling has been implicated in autoimmune and inflammatory disorders. However, the molecular mechanism by which the OX40 cosignaling regulates the T-cell response remains obscure. We found that OX40 associated with a scaffold protein, IQ motif-containing GTPase-activating protein 1 (IQGAP1) after ligation by its ligand OX40L. Naïve CD4
+ T cells from Iqgap1-/- mice displayed enhanced proliferation and cytokine secretion upon receiving OX40 cosignaling. A C-terminal IQGAP1 region was responsible for its association with OX40, and TNFR-associated factor 2 (TRAF2) bridged these two proteins. The enhanced cytokine response in Iqgap1-/- T cells was restored by the expression of the C-terminal IQGAP1. Thus, the IQGAP1 binding limits the OX40 cosignaling. Disease severity of experimental autoimmune encephalomyelitis (EAE) was significantly exacerbated in Iqgap1-/- mice as compared to wild-type mice. Additionally, recipient mice with Iqgap1-/- donor CD4+ T cells exhibited significantly higher EAE scores than those with their wild-type counterparts, and OX40 blockade led to a significant reduction in the EAE severity. Thus, our study defines an important component of the OX40 cosignaling that restricts inflammation driven by antigen-activated T cells., (© 2019 The Authors. The FASEB Journal published by Wiley Periodicals, Inc. on behalf of Federation of American Societies for Experimental Biology.)- Published
- 2020
- Full Text
- View/download PDF
36. Novel interaction of antioxidant-1 with TRAF4: role in inflammatory responses in endothelial cells.
- Author
-
Das A, Sudhahar V, Ushio-Fukai M, and Fukai T
- Subjects
- Angiotensin II administration & dosage, Animals, Aorta metabolism, Aorta pathology, Apolipoproteins E deficiency, Apolipoproteins E genetics, Atherosclerosis etiology, Atherosclerosis metabolism, Atherosclerosis pathology, Copper metabolism, Copper Transport Proteins metabolism, Diet, High-Fat adverse effects, Gene Expression Regulation, HEK293 Cells, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells metabolism, Humans, Inflammation, Intercellular Adhesion Molecule-1 genetics, Intercellular Adhesion Molecule-1 metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, ApoE, Molecular Chaperones metabolism, NADPH Oxidases metabolism, Protein Binding, Protein Transport drug effects, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, TNF Receptor-Associated Factor 4 antagonists & inhibitors, TNF Receptor-Associated Factor 4 metabolism, Tumor Necrosis Factor-alpha pharmacology, Vascular Cell Adhesion Molecule-1 genetics, Vascular Cell Adhesion Molecule-1 metabolism, Atherosclerosis genetics, Copper Transport Proteins genetics, Molecular Chaperones genetics, NADPH Oxidases genetics, Reactive Oxygen Species metabolism, TNF Receptor-Associated Factor 4 genetics
- Abstract
NADPH oxidase (NOX)-derived reactive oxygen species (ROS) and copper (Cu), an essential micronutrient, have been implicated in vascular inflammatory diseases. We reported that in proinflammatory cytokine TNF-α-stimulated endothelial cells (ECs), cytosolic Cu chaperone antioxidant-1 (Atox1) functions as a Cu-dependent transcription factor for the NOX organizer p47phox, thereby increasing ROS-dependent inflammatory gene expression. However, the role and mechanism of Atox1 nuclear translocation in inflamed ECs remain unclear. Using enface staining and nuclear fractionation, here we show that Atox1 was localized in the nucleus in inflamed aortas from ApoE
-/- mice with angiotensin II infusion on a high-fat diet, while it was found in cytosol in those from control mice. In cultured human ECs, TNF-α stimulation promoted Atox1 nuclear translocation within 15 min, which was associated with Atox1 binding to TNF-α receptor-associated factor 4 (TRAF4) in a Cu-dependent manner. TRAF4 depletion by siRNA significantly inhibited Atox1 nuclear translocation, p47phox expression, and ROS production as well as its downstream VCAM1/ICAM1 expression and monocyte adhesion to inflamed ECs, which were rescued by overexpression of nuclear targeted Atox1. Furthermore, Atox1 colocalized with TRAF4 at the nucleus in TNF-α-stimulated inflamed ECs and vessels. In summary, Cu-dependent Atox1 binding to TRAF4 plays an important role in Atox1 nuclear translocation and ROS-dependent inflammatory responses in TNF-α-stimulated ECs. Thus the Atox1-TRAF4 axis is a novel therapeutic target for vascular inflammatory disease such as atherosclerosis.- Published
- 2019
- Full Text
- View/download PDF
37. Copper Transporter ATP7A (Copper-Transporting P-Type ATPase/Menkes ATPase) Limits Vascular Inflammation and Aortic Aneurysm Development: Role of MicroRNA-125b.
- Author
-
Sudhahar V, Das A, Horimatsu T, Ash D, Leanhart S, Antipova O, Vogt S, Singla B, Csanyi G, White J, Kaplan JH, Fulton D, Weintraub NL, Kim HW, Ushio-Fukai M, and Fukai T
- Subjects
- Angiotensin II drug effects, Animals, Apoptosis, Cells, Cultured, Chelating Agents pharmacology, Copper metabolism, Copper Transport Proteins metabolism, Copper-Transporting ATPases genetics, Disease Models, Animal, Down-Regulation, Female, Humans, Inflammation genetics, Inflammation physiopathology, Male, Mice, Inbred C57BL, Mice, Transgenic, Molecular Chaperones metabolism, Molybdenum pharmacology, Muscle, Smooth, Vascular cytology, Up-Regulation, Aortic Aneurysm, Abdominal genetics, Aortic Aneurysm, Abdominal physiopathology, Copper-Transporting ATPases physiology, MicroRNAs physiology
- Abstract
Objective: Copper (Cu) is essential micronutrient, and its dysregulation is implicated in aortic aneurysm (AA) development. The Cu exporter ATP7A (copper-transporting P-type ATPase/Menkes ATPase) delivers Cu via the Cu chaperone Atox1 (antioxidant 1) to secretory Cu enzymes, such as lysyl oxidase, and excludes excess Cu. Lysyl oxidase is shown to protect against AA formation. However, the role and mechanism of ATP7A in AA pathogenesis remain unknown. Approach and Results: Here, we show that Cu chelator markedly inhibited Ang II (angiotensin II)-induced abdominal AA (AAA) in which ATP7A expression was markedly downregulated. Transgenic ATP7A overexpression prevented Ang II-induced AAA formation. Conversely, Cu transport dysfunctional ATP7A
mut/+ /ApoE-/- mice exhibited robust AAA formation and dissection, excess aortic Cu accumulation as assessed by X-ray fluorescence microscopy, and reduced lysyl oxidase activity. In contrast, AAA formation was not observed in Atox1-/- /ApoE-/- mice, suggesting that decreased lysyl oxidase activity, which depends on both ATP7A and Atox1, was not sufficient to develop AAA. Bone marrow transplantation suggested importance of ATP7A in vascular cells, not bone marrow cells, in AAA development. MicroRNA (miR) array identified miR-125b as a highly upregulated miR in AAA from ATP7Amut/+ /ApoE-/- mice. Furthermore, miR-125b target genes (histone methyltransferase Suv39h1 and the NF-κB negative regulator TNFAIP3 [tumor necrosis factor alpha induced protein 3]) were downregulated, which resulted in increased proinflammatory cytokine expression, aortic macrophage recruitment, MMP (matrix metalloproteinase)-2/9 activity, elastin fragmentation, and vascular smooth muscle cell loss in ATP7Amut/+ /ApoE-/- mice and reversed by locked nucleic acid-anti-miR-125b infusion., Conclusions: ATP7A downregulation/dysfunction promotes AAA formation via upregulating miR-125b, which augments proinflammatory signaling in a Cu-dependent manner. Thus, ATP7A is a potential therapeutic target for inflammatory vascular disease.- Published
- 2019
- Full Text
- View/download PDF
38. Modification of Cardiac Progenitor Cell-Derived Exosomes by miR-322 Provides Protection against Myocardial Infarction through Nox2-Dependent Angiogenesis.
- Author
-
Youn SW, Li Y, Kim YM, Sudhahar V, Abdelsaid K, Kim HW, Liu Y, Fulton DJR, Ashraf M, Tang Y, Fukai T, and Ushio-Fukai M
- Abstract
Myocardial infarction (MI) is the primary cause of cardiovascular mortality, and therapeutic strategies to prevent or mitigate the consequences of MI are a high priority. Cardiac progenitor cells (CPCs) have been used to treat cardiac injury post-MI, and despite poor engraftment, they have been shown to inhibit apoptosis and to promote angiogenesis through poorly understood paracrine effects. We previously reported that the direct injection of exosomes derived from CPCs (CPCexo) into mouse hearts provides protection against apoptosis in a model of acute ischemia/reperfusion injury. Moreover, we and others have reported that reactive oxygen species (ROS) derived from NADPH oxidase (NOX) can enhance angiogenesis in endothelial cells (ECs). Here we examined whether bioengineered CPCexo transfected with a pro-angiogenic miR-322 (CPCexo-322) can improve therapeutic efficacy in a mouse model of MI as compared to CPCexo. Systemic administration of CPCexo-322 in mice after ischemic injury provided greater protection post-MI than control CPCexo, in part, through enhanced angiogenesis in the border zones of infarcted hearts. Mechanistically, the treatment of cultured human ECs with CPCexo-322 resulted in a greater angiogenic response, as determined by increased EC migration and capillary tube formation via increased Nox2-derived ROS. Our study reveals that the engineering of CPCexo via microRNA (miR) programing can enhance angiogenesis, and this may be an effective therapeutic strategy for the treatment of ischemic cardiovascular diseases.
- Published
- 2019
- Full Text
- View/download PDF
39. Copper transporter ATP7A interacts with IQGAP1, a Rac1 binding scaffolding protein: role in PDGF-induced VSMC migration and vascular remodeling.
- Author
-
Ashino T, Kohno T, Sudhahar V, Ash D, Ushio-Fukai M, and Fukai T
- Subjects
- Animals, Aorta cytology, Aorta metabolism, Cell Movement genetics, Copper chemistry, Copper metabolism, Gene Expression Regulation, Developmental genetics, Membrane Microdomains genetics, Membrane Microdomains metabolism, Mice, Muscle, Smooth, Vascular growth & development, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Neointima genetics, Phosphorylation, Protein Binding, Rats, Copper-Transporting ATPases genetics, Platelet-Derived Growth Factor genetics, Vascular Remodeling genetics, rac1 GTP-Binding Protein genetics, ras GTPase-Activating Proteins genetics
- Abstract
Vascular smooth muscle cell (VSMC) migration contributes to neointimal formation after vascular injury. We previously demonstrated that copper (Cu) transporter ATP7A is involved in platelet-derived growth factor (PDGF)-induced VSMC migration in a Cu- and Rac1-dependent manner. The underlying mechanism is still unknown. Here we show that ATP7A interacts with IQGAP1, a Rac1 and receptor tyrosine kinase binding scaffolding proteins, which mediates PDGF-induced VSMC migration and vascular remodeling. In cultured rat aortic SMCs, PDGF stimulation rapidly promoted ATP7A association with IQGAP1 and Rac1 and their translocation to the lipid rafts and leading edge. Cotransfection assay revealed that ATP7A directly bound to NH
2 -terminal domain of IQGAP1. Functionally, either ATP7A or IQGAP1 depletion using siRNA significantly inhibited PDGF-induced VSMC migration without additive effects, suggesting that IQGAP1 and ATP7A are in the same axis to promote migration. Furthermore, IQGAP1 siRNA blocked PDGF-induced ATP7A association with Rac1 as well as its translocation to leading edge, while PDGF-induced IQGAP1 translocation was not affected by ATP7A siRNA or Cu chelator. Overexpression of mutant IQGAP1 lacking a Rac1 binding site prevented PDGF-induced translocation of Rac1, but not ATP7A, to the leading edge, thereby inhibiting lamellipodia formation and VSMC migration. In vivo, ATP7A colocalized with IQGAP1 at neointimal VSMCs in a mice wire injury model, while neointimal formation and extracellular matrix deposition induced by vascular injury were inhibited in ATP7A mutant mice with reduced Cu transporter function. In summary, IQGAP1 functions as ATP7A and Rac1 binding scaffolding protein to organize PDGF-dependent ATP7A translocation to the lamellipodial leading edge, thereby promoting VSMC migration and vascular remodeling.- Published
- 2018
- Full Text
- View/download PDF
40. Oxidant Signaling Mediated by Nox2 in Neutrophils Promotes Regenerative Myelopoiesis and Tissue Recovery following Ischemic Damage.
- Author
-
Fang MM, Barman PK, Thiruppathi M, Mirza RE, McKinney RD, Deng J, Christman JW, Du X, Fukai T, Ennis WJ, Koh TJ, Ushio-Fukai M, and Urao N
- Subjects
- Animals, Cells, Cultured, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Myelopoiesis, NADPH Oxidase 2 genetics, Oxidation-Reduction, Reactive Oxygen Species metabolism, Regeneration, Signal Transduction, src-Family Kinases metabolism, Hematopoietic Stem Cells physiology, Hindlimb pathology, Ischemia immunology, NADPH Oxidase 2 metabolism, Neutrophils physiology
- Abstract
Ischemic tissue damage activates hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM)-generating myeloid cells, and persistent HSPC activity may drive chronic inflammation and impair tissue recovery. Although increased reactive oxygen species in the BM regulate HSPC functions, their roles in myelopoiesis of activated HSPCs and subsequent tissue recovery during ischemic damage are not well understood. In this paper, we report that deletion of Nox2 NADPH oxidase in mice results in persistent elevations in BM HSPC activity and levels of inflammatory monocytes/macrophages in BM and ischemic tissue in a model of hindlimb ischemia. Ischemic tissue damage induces oxidants in BM such as elevations of hydrogen peroxide and oxidized phospholipids, which activate redox-sensitive Lyn kinase in a Nox2-dependent manner. Moreover, during tissue recovery after ischemic injury, this Nox2-ROS-Lyn kinase axis is induced by Nox2 in neutrophils that home to the BM, which inhibits HSPC activity and inflammatory monocyte generation and promotes tissue regeneration after ischemic damage. Thus, oxidant signaling in the BM mediated by Nox2 in neutrophils regulates myelopoiesis of HSPCs to promote regeneration of damaged tissue., (Copyright © 2018 by The American Association of Immunologists, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
41. Copper transporters and copper chaperones: roles in cardiovascular physiology and disease.
- Author
-
Fukai T, Ushio-Fukai M, and Kaplan JH
- Subjects
- Animals, Cardiovascular Diseases pathology, Cardiovascular System physiopathology, Gene Expression physiology, Humans, Signal Transduction physiology, Cardiovascular Diseases metabolism, Cardiovascular System metabolism, Cation Transport Proteins metabolism, Copper metabolism, Molecular Chaperones metabolism
- Abstract
Copper (Cu) is an essential micronutrient but excess Cu is potentially toxic. Its important propensity to cycle between two oxidation states accounts for its frequent presence as a cofactor in many physiological processes through Cu-containing enzymes, including mitochondrial energy production (via cytochrome c-oxidase), protection against oxidative stress (via superoxide dismutase), and extracellular matrix stability (via lysyl oxidase). Since free Cu is potentially toxic, the bioavailability of intracellular Cu is tightly controlled by Cu transporters and Cu chaperones. Recent evidence reveals that these Cu transport systems play an essential role in the physiological responses of cardiovascular cells, including cell growth, migration, angiogenesis and wound repair. In response to growth factors, cytokines, and hypoxia, their expression, subcellular localization, and function are tightly regulated. Cu transport systems and their regulators have also been linked to various cardiovascular pathophysiologies such as hypertension, inflammation, atherosclerosis, diabetes, cardiac hypertrophy, and cardiomyopathy. A greater appreciation of the central importance of Cu transporters and Cu chaperones in cell signaling and gene expression in cardiovascular biology offers the possibility of identifying new therapeutic targets for cardiovascular disease.
- Published
- 2018
- Full Text
- View/download PDF
42. Redox Regulation of Mitochondrial Fission Protein Drp1 by Protein Disulfide Isomerase Limits Endothelial Senescence.
- Author
-
Kim YM, Youn SW, Sudhahar V, Das A, Chandhri R, Cuervo Grajal H, Kweon J, Leanhart S, He L, Toth PT, Kitajewski J, Rehman J, Yoon Y, Cho J, Fukai T, and Ushio-Fukai M
- Subjects
- Animals, Cell Respiration, Cysteine metabolism, Diabetes Mellitus, Type 2 pathology, Endoplasmic Reticulum Stress, Humans, Mice, Mitochondria metabolism, Mutation genetics, Oxidation-Reduction, Protein Binding, Reactive Oxygen Species metabolism, Wound Healing, Cellular Senescence, Dynamins metabolism, Human Umbilical Vein Endothelial Cells metabolism, Mitochondrial Dynamics, Procollagen-Proline Dioxygenase metabolism, Protein Disulfide-Isomerases metabolism
- Abstract
Mitochondrial dynamics are tightly controlled by fusion and fission, and their dysregulation and excess reactive oxygen species (ROS) contribute to endothelial cell (EC) dysfunction. How redox signals regulate coupling between mitochondrial dynamics and endothelial (dys)function remains unknown. Here, we identify protein disulfide isomerase A1 (PDIA1) as a thiol reductase for the mitochondrial fission protein Drp1. A biotin-labeled Cys-OH trapping probe and rescue experiments reveal that PDIA1 depletion in ECs induces sulfenylation of Drp1 at Cys
644 , promoting mitochondrial fragmentation and ROS elevation without inducing ER stress, which drives EC senescence. Mechanistically, PDIA1 associates with Drp1 to reduce its redox status and activity. Defective wound healing and angiogenesis in diabetic or PDIA1+/- mice are restored by EC-targeted PDIA1 or the Cys oxidation-defective mutant Drp1. Thus, this study uncovers a molecular link between PDIA1 and Drp1 oxidoreduction, which maintains normal mitochondrial dynamics and limits endothelial senescence with potential translational implications for vascular diseases associated with diabetes or aging., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
43. Akt2 (Protein Kinase B Beta) Stabilizes ATP7A, a Copper Transporter for Extracellular Superoxide Dismutase, in Vascular Smooth Muscle: Novel Mechanism to Limit Endothelial Dysfunction in Type 2 Diabetes Mellitus.
- Author
-
Sudhahar V, Okur MN, Bagi Z, O'Bryan JP, Hay N, Makino A, Patel VS, Phillips SA, Stepp D, Ushio-Fukai M, and Fukai T
- Subjects
- Animals, Aorta, Thoracic enzymology, Aorta, Thoracic physiopathology, Cells, Cultured, Copper-Transporting ATPases genetics, Diabetes Mellitus, Experimental drug therapy, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental physiopathology, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 physiopathology, Diabetic Angiopathies genetics, Diabetic Angiopathies physiopathology, Diabetic Angiopathies prevention & control, Endothelium, Vascular drug effects, Endothelium, Vascular physiopathology, Enzyme Stability, Female, Humans, Hypoglycemic Agents pharmacology, Insulin pharmacology, Male, Mesenteric Arteries enzymology, Mesenteric Arteries physiopathology, Mice, Inbred C57BL, Mice, Knockout, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular physiopathology, Phosphorylation, Protein Transport, Proto-Oncogene Proteins c-akt deficiency, Proto-Oncogene Proteins c-akt genetics, Rats, Sprague-Dawley, Signal Transduction, Superoxide Dismutase deficiency, Superoxide Dismutase genetics, Vasodilation, Copper-Transporting ATPases metabolism, Diabetes Mellitus, Experimental enzymology, Diabetes Mellitus, Type 2 enzymology, Diabetic Angiopathies enzymology, Endothelium, Vascular enzymology, Muscle, Smooth, Vascular enzymology, Proto-Oncogene Proteins c-akt metabolism, Superoxide Dismutase metabolism
- Abstract
Objective: Copper transporter ATP7A (copper-transporting/ATPase) is required for full activation of SOD3 (extracellular superoxide dismutase), which is secreted from vascular smooth muscle cells (VSMCs) and anchors to endothelial cell surface to preserve endothelial function by scavenging extracellular superoxide. We reported that ATP7A protein expression and SOD3 activity are decreased in insulin-deficient type 1 diabetes mellitus vessels, thereby, inducing superoxide-mediated endothelial dysfunction, which are rescued by insulin treatment. However, it is unknown regarding the mechanism by which insulin increases ATP7A expression in VSMCs and whether ATP7A downregulation is observed in T2DM (type2 diabetes mellitus) mice and human in which insulin-Akt (protein kinase B) pathway is selectively impaired., Approach and Results: Here we show that ATP7A protein is markedly downregulated in vessels isolated from T2DM patients, as well as those from high-fat diet-induced or db/db T2DM mice. Akt2 (protein kinase B beta) activated by insulin promotes ATP7A stabilization via preventing ubiquitination/degradation as well as translocation to plasma membrane in VSMCs, which contributes to activation of SOD3 that protects against T2DM-induced endothelial dysfunction. Downregulation of ATP7A in T2DM vessels is restored by constitutive active Akt or PTP1B
-/- (protein-tyrosine phosphatase 1B-deficient) T2DM mice, which enhance insulin-Akt signaling. Immunoprecipitation, in vitro kinase assay, and mass spectrometry analysis reveal that insulin stimulates Akt2 binding to ATP7A to induce phosphorylation at Ser1424/1463/1466. Furthermore, SOD3 activity is reduced in Akt2-/- vessels or VSMCs, which is rescued by ATP7A overexpression., Conclusion: Akt2 plays a critical role in ATP7A protein stabilization and translocation to plasma membrane in VSMCs, which contributes to full activation of vascular SOD3 that protects against endothelial dysfunction in T2DM., (© 2018 American Heart Association, Inc.)- Published
- 2018
- Full Text
- View/download PDF
44. Embryonic Stem Cell Differentiation to Functional Arterial Endothelial Cells through Sequential Activation of ETV2 and NOTCH1 Signaling by HIF1α.
- Author
-
Tsang KM, Hyun JS, Cheng KT, Vargas M, Mehta D, Ushio-Fukai M, Zou L, Pajcini KV, Rehman J, and Malik AB
- Subjects
- Animals, Cell Hypoxia, Cell Lineage, Endothelial Cells transplantation, Hindlimb pathology, Ischemia pathology, Mice, Mice, Inbred C57BL, Mouse Embryonic Stem Cells metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Organogenesis, Signal Transduction, Transcription Factors genetics, Up-Regulation genetics, Arteries cytology, Cell Differentiation, Endothelial Cells cytology, Endothelial Cells metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mouse Embryonic Stem Cells cytology, Receptors, Notch metabolism, Transcription Factors metabolism
- Abstract
The generation of functional arterial endothelial cells (aECs) from embryonic stem cells (ESCs) holds great promise for vascular tissue engineering. However, the mechanisms underlying their generation and the potential of aECs in revascularizing ischemic tissue are not fully understood. Here, we observed that hypoxia exposure of mouse ESCs induced an initial phase of HIF1α-mediated upregulation of the transcription factor Etv2, which in turn induced the commitment to the EC fate. However, sustained activation of HIF1α in these EC progenitors thereafter induced NOTCH1 signaling that promoted the transition to aEC fate. We observed that transplantation of aECs mediated arteriogenesis in the mouse hindlimb ischemia model. Furthermore, transplantation of aECs in mice showed engraftment in ischemic myocardium and restored cardiac function in contrast to ECs derived under normoxia. Thus, HIF1α activation of Etv2 in ESCs followed by NOTCH1 signaling is required for the generation aECs that are capable of arteriogenesis and revascularization of ischemic tissue., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
45. ROS-induced ROS release orchestrated by Nox4, Nox2, and mitochondria in VEGF signaling and angiogenesis.
- Author
-
Kim YM, Kim SJ, Tatsunami R, Yamamura H, Fukai T, and Ushio-Fukai M
- Subjects
- Biosensing Techniques, Catalase genetics, Catalase metabolism, Cell Movement drug effects, Cell Proliferation drug effects, Gene Expression Regulation, Human Umbilical Vein Endothelial Cells, Humans, Membrane Glycoproteins antagonists & inhibitors, Membrane Glycoproteins metabolism, Microscopy, Fluorescence, Mitochondria drug effects, NADPH Oxidase 2, NADPH Oxidase 4, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases metabolism, Neovascularization, Physiologic drug effects, Neovascularization, Physiologic genetics, Oxidation-Reduction, Phosphorylation drug effects, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Src Homology 2 Domain-Containing, Transforming Protein 1 genetics, Src Homology 2 Domain-Containing, Transforming Protein 1 metabolism, Time-Lapse Imaging, Vascular Endothelial Growth Factor A pharmacology, Vascular Endothelial Growth Factor Receptor-2 genetics, Vascular Endothelial Growth Factor Receptor-2 metabolism, Feedback, Physiological, Hydrogen Peroxide metabolism, Membrane Glycoproteins genetics, Mitochondria metabolism, NADPH Oxidases genetics, Signal Transduction
- Abstract
Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) and mitochondria play a critical role in growth factor-induced switch from a quiescent to an angiogenic phenotype in endothelial cells (ECs). However, how highly diffusible ROS produced from different sources can coordinate to stimulate VEGF signaling and drive the angiogenic process remains unknown. Using the cytosol- and mitochondria-targeted redox-sensitive RoGFP biosensors with real-time imaging, here we show that VEGF stimulation in human ECs rapidly increases cytosolic RoGFP oxidation within 1 min, followed by mitochondrial RoGFP oxidation within 5 min, which continues at least for 60 min. Silencing of Nox4 or Nox2 or overexpression of mitochondria-targeted catalase significantly inhibits VEGF-induced tyrosine phosphorylation of VEGF receptor type 2 (VEGFR2-pY), EC migration and proliferation at the similar extent. Exogenous hydrogen peroxide (H
2 O2 ) or overexpression of Nox4, which produces H2 O2 , increases mitochondrial ROS (mtROS), which is prevented by Nox2 siRNA, suggesting that Nox2 senses Nox4-derived H2 O2 to promote mtROS production. Mechanistically, H2 O2 increases S36 phosphorylation of p66Shc, a key mtROS regulator, which is inhibited by siNox2, but not by siNox4. Moreover, Nox2 or Nox4 knockdown or overexpression of S36 phosphorylation-defective mutant p66Shc(S36A) inhibits VEGF-induced mtROS, VEGFR2-pY, EC migration, and proliferation. In summary, Nox4-derived H2 O2 in part activates Nox2 to increase mtROS via pSer36-p66Shc, thereby enhancing VEGFR2 signaling and angiogenesis in ECs. This may represent a novel feed-forward mechanism of ROS-induced ROS release orchestrated by the Nox4/Nox2/pSer36-p66Shc/mtROS axis, which drives sustained activation of angiogenesis signaling program., (Copyright © 2017 the American Physiological Society.)- Published
- 2017
- Full Text
- View/download PDF
46. Short-term regular aerobic exercise reduces oxidative stress produced by acute in the adipose microvasculature.
- Author
-
Robinson AT, Fancher IS, Sudhahar V, Bian JT, Cook MD, Mahmoud AM, Ali MM, Ushio-Fukai M, Brown MD, Fukai T, and Phillips SA
- Subjects
- Angiotensin II Type 2 Receptor Blockers pharmacology, Animals, Arteries physiology, Blood Pressure physiology, Endothelial Cells drug effects, Hindlimb blood supply, Humans, Male, Membrane Glycoproteins antagonists & inhibitors, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, NADPH Oxidase 2, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases genetics, Superoxide Dismutase antagonists & inhibitors, Superoxide Dismutase genetics, Vascular Resistance, Adipose Tissue blood supply, Adipose Tissue physiology, Exercise physiology, Microvessels physiology, Oxidative Stress physiology
- Abstract
High blood pressure has been shown to elicit impaired dilation in the vasculature. The purpose of this investigation was to elucidate the mechanisms through which high pressure may elicit vascular dysfunction and determine the mechanisms through which regular aerobic exercise protects arteries against high pressure. Male C57BL/6J mice were subjected to 2 wk of voluntary running (~6 km/day) for comparison with sedentary controls. Hindlimb adipose resistance arteries were dissected from mice for measurements of flow-induced dilation (FID; with or without high intraluminal pressure exposure) or protein expression of NADPH oxidase II (NOX II) and superoxide dismutase (SOD). Microvascular endothelial cells were subjected to high physiological laminar shear stress (20 dyn/cm
2 ) or static condition and treated with ANG II + pharmacological inhibitors. Cells were analyzed for the detection of ROS or collected for Western blot determination of NOX II and SOD. Resistance arteries from exercised mice demonstrated preserved FID after high pressure exposure, whereas FID was impaired in control mouse arteries. Inhibition of ANG II or NOX II restored impaired FID in control mouse arteries. High pressure increased superoxide levels in control mouse arteries but not in exercise mouse arteries, which exhibited greater ability to convert superoxide to H2 O2 Arteries from exercised mice exhibited less NOX II protein expression, more SOD isoform expression, and less sensitivity to ANG II. Endothelial cells subjected to laminar shear stress exhibited less NOX II subunit expression. In conclusion, aerobic exercise prevents high pressure-induced vascular dysfunction through an improved redox environment in the adipose microvasculature. NEW & NOTEWORTHY We describe potential mechanisms contributing to aerobic exercise-conferred protection against high intravascular pressure. Subcutaneous adipose microvessels from exercise mice express less NADPH oxidase (NOX) II and more superoxide dismutase (SOD) and demonstrate less sensitivity to ANG II. In microvascular endothelial cells, shear stress reduced NOX II but did not influence SOD expression.- Published
- 2017
- Full Text
- View/download PDF
47. Endothelial Antioxidant-1: a Key Mediator of Copper-dependent Wound Healing in vivo.
- Author
-
Das A, Sudhahar V, Chen GF, Kim HW, Youn SW, Finney L, Vogt S, Yang J, Kweon J, Surenkhuu B, Ushio-Fukai M, and Fukai T
- Abstract
Copper (Cu), an essential nutrient, promotes wound healing, however, target of Cu action and underlying mechanisms remain elusive. Cu chaperone Antioxidant-1 (Atox1) in the cytosol supplies Cu to the secretory enzymes such as lysyl oxidase (LOX), while Atox1 in the nucleus functions as a Cu-dependent transcription factor. Using mouse cutaneous wound healing model, here we show that Cu content (by X-ray Fluorescence Microscopy) and nuclear Atox1 are increased after wounding, and that wound healing with and without Cu treatment is impaired in Atox1
-/- mice. Endothelial cell (EC)-specific Atox1-/- mice and gene transfer of nuclear-target Atox1 in Atox1-/- mice reveal that Atox1 in ECs as well as transcription factor function of Atox1 are required for wound healing. Mechanistically, Atox1-/- mice show reduced Atox1 target proteins such as p47phox NADPH oxidase and cyclin D1 as well as extracellular matrix Cu enzyme LOX activity in wound tissues. This in turn results in reducing O2 - production in ECs, NFkB activity, cell proliferation and collagen formation, thereby inhibiting angiogenesis, macrophage recruitment and extracellular matrix maturation. Our findings suggest that Cu-dependent transcription factor/Cu chaperone Atox1 in ECs plays an important role to sense Cu to accelerate wound angiogenesis and healing.- Published
- 2016
- Full Text
- View/download PDF
48. Differential Roles of the NADPH-Oxidase 1 and 2 in Platelet Activation and Thrombosis.
- Author
-
Delaney MK, Kim K, Estevez B, Xu Z, Stojanovic-Terpo A, Shen B, Ushio-Fukai M, Cho J, and Du X
- Subjects
- Animals, Blood Platelets drug effects, CD11b Antigen blood, Calcium Signaling, Disease Models, Animal, Enzyme Activation, Genetic Predisposition to Disease, Hemostasis, Male, Membrane Glycoproteins deficiency, Membrane Glycoproteins genetics, Mice, Inbred C57BL, Mice, Knockout, Mitogen-Activated Protein Kinases metabolism, NADH, NADPH Oxidoreductases deficiency, NADH, NADPH Oxidoreductases genetics, NADPH Oxidase 1, NADPH Oxidase 2, NADPH Oxidases deficiency, NADPH Oxidases genetics, Phenotype, Phospholipase C gamma blood, Phosphorylation, Platelet Aggregation, Platelet Membrane Glycoproteins agonists, Platelet Membrane Glycoproteins metabolism, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled blood, Syk Kinase blood, Thrombin metabolism, Thrombosis genetics, Time Factors, Blood Platelets enzymology, Membrane Glycoproteins blood, NADH, NADPH Oxidoreductases blood, NADPH Oxidases blood, Platelet Activation drug effects, Reactive Oxygen Species blood, Thrombosis blood, Thrombosis enzymology
- Abstract
Objective: Reactive oxygen species (ROS) are known to regulate platelet activation; however, the mechanisms of ROS production during platelet activation remain unclear. Platelets express different isoforms of nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidases (NOXs). Here, we investigated the role of NOX1 and NOX2 in ROS generation and platelet activation using NOX1 and NOX2 knockout mice., Approach and Results: NOX1(-/Y) platelets showed selective defects in G-protein-coupled receptor-mediated platelet activation induced by thrombin and thromboxane A2 analog U46619, but were not affected in platelet activation induced by collagen-related peptide, a glycoprotein VI agonist. In contrast, NOX2(-/-) platelets showed potent inhibition of collagen-related peptide-induced platelet activation, and also showed partial inhibition of thrombin-induced platelet activation. Consistently, production of ROS was inhibited in NOX1(-/Y) platelets stimulated with thrombin, but not collagen-related peptide, whereas NOX2(-/-) platelets showed reduced ROS generation induced by collagen-related peptide or thrombin. Reduced ROS generation in NOX1/2-deficient platelets is associated with impaired activation of Syk and phospholipase Cγ2, but minimally affected mitogen-activated protein kinase pathways. Interestingly, laser-induced arterial thrombosis was impaired but the bleeding time was not affected in NOX2(-/-) mice. Wild-type thrombocytopenic mice injected with NOX2(-/-) platelets also showed defective arterial thrombosis, suggesting an important role for platelet NOX2 in thrombosis in vivo but not hemostasis., Conclusions: NOX1 and NOX2 play differential roles in different platelet activation pathways and in thrombosis. ROS generated by these enzymes promotes platelet activation via the Syk/phospholipase Cγ2/calcium signaling pathway., (© 2016 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
49. Injury-Mediated Vascular Regeneration Requires Endothelial ER71/ETV2.
- Author
-
Park C, Lee TJ, Bhang SH, Liu F, Nakamura R, Oladipupo SS, Pitha-Rowe I, Capoccia B, Choi HS, Kim TM, Urao N, Ushio-Fukai M, Lee DJ, Miyoshi H, Kim BS, Lim DS, Apte RS, Ornitz DM, and Choi K
- Subjects
- Angiogenic Proteins deficiency, Angiogenic Proteins genetics, Animals, Cells, Cultured, Choroidal Neovascularization genetics, Choroidal Neovascularization metabolism, Choroidal Neovascularization physiopathology, Disease Models, Animal, Endothelial Cells pathology, Gene Expression Regulation, Gene Transfer Techniques, Genetic Vectors, Heterozygote, Hindlimb, Ischemia genetics, Ischemia pathology, Ischemia physiopathology, Ischemia therapy, Lentivirus genetics, Male, Mice, Inbred C57BL, Mice, Knockout, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Phenotype, Recovery of Function, Signal Transduction, Skin blood supply, Time Factors, Transcription Factors deficiency, Transcription Factors genetics, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor Receptor-2 genetics, Vascular Endothelial Growth Factor Receptor-2 metabolism, Wound Healing, Angiogenic Proteins metabolism, Endothelial Cells metabolism, Ischemia metabolism, Muscle, Skeletal blood supply, Neovascularization, Physiologic, Regeneration, Transcription Factors metabolism
- Abstract
Objective: Comprehensive understanding of the mechanisms regulating angiogenesis might provide new strategies for angiogenic therapies for treating diverse physiological and pathological ischemic conditions. The E-twenty six (ETS) factor Ets variant 2 (ETV2; aka Ets-related protein 71) is essential for the formation of hematopoietic and vascular systems. Despite its indispensable function in vessel development, ETV2 role in adult angiogenesis has not yet been addressed. We have therefore investigated the role of ETV2 in vascular regeneration., Approach and Results: We used endothelial Etv2 conditional knockout mice and ischemic injury models to assess the role of ETV2 in vascular regeneration. Although Etv2 expression was not detectable under steady-state conditions, its expression was readily observed in endothelial cells after injury. Mice lacking endothelial Etv2 displayed impaired neovascularization in response to eye injury, wounding, or hindlimb ischemic injury. Lentiviral Etv2 expression in ischemic hindlimbs led to improved recovery of blood perfusion with enhanced vessel formation. After injury, fetal liver kinase 1 (Flk1), aka VEGFR2, expression and neovascularization were significantly upregulated by Etv2, whereas Flk1 expression and vascular endothelial growth factor response were significantly blunted in Etv2-deficient endothelial cells. Conversely, enforced Etv2 expression enhanced vascular endothelial growth factor-mediated endothelial sprouting from embryoid bodies. Lentiviral Flk1 expression rescued angiogenesis defects in endothelial Etv2 conditional knockout mice after hindlimb ischemic injury. Furthermore, Etv2(+/-); Flk1(+/-) double heterozygous mice displayed a more severe hindlimb ischemic injury response compared with Etv2(+/-) or Flk1(+/-) heterozygous mice, revealing an epistatic interaction between ETV2 and FLK1 in vascular regeneration., Conclusions: Our study demonstrates a novel obligatory role for the ETV2 in postnatal vascular repair and regeneration., (© 2015 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
50. Copper Transport Protein Antioxidant-1 Promotes Inflammatory Neovascularization via Chaperone and Transcription Factor Function.
- Author
-
Chen GF, Sudhahar V, Youn SW, Das A, Cho J, Kamiya T, Urao N, McKinney RD, Surenkhuu B, Hamakubo T, Iwanari H, Li S, Christman JW, Shantikumar S, Angelini GD, Emanueli C, Ushio-Fukai M, and Fukai T
- Subjects
- Adenosine Triphosphatases metabolism, Animals, Cation Transport Proteins metabolism, Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Cell Line, Copper Transport Proteins, Copper-Transporting ATPases, Gene Expression Regulation, Hindlimb, Human Umbilical Vein Endothelial Cells cytology, Humans, Ischemia metabolism, Ischemia pathology, Leg blood supply, Leg pathology, Metallochaperones metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Molecular Chaperones, Monocytes metabolism, Monocytes pathology, NADPH Oxidases metabolism, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Protein-Lysine 6-Oxidase metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor A metabolism, Adenosine Triphosphatases genetics, Cation Transport Proteins genetics, Human Umbilical Vein Endothelial Cells metabolism, Ischemia genetics, Metallochaperones genetics, NADPH Oxidases genetics, Neovascularization, Pathologic genetics, Protein-Lysine 6-Oxidase genetics
- Abstract
Copper (Cu), an essential micronutrient, plays a fundamental role in inflammation and angiogenesis; however, its precise mechanism remains undefined. Here we uncover a novel role of Cu transport protein Antioxidant-1 (Atox1), which is originally appreciated as a Cu chaperone and recently discovered as a Cu-dependent transcription factor, in inflammatory neovascularization. Atox1 expression is upregulated in patients and mice with critical limb ischemia. Atox1-deficient mice show impaired limb perfusion recovery with reduced arteriogenesis, angiogenesis, and recruitment of inflammatory cells. In vivo intravital microscopy, bone marrow reconstitution, and Atox1 gene transfer in Atox1(-/-) mice show that Atox1 in endothelial cells (ECs) is essential for neovascularization and recruitment of inflammatory cells which release VEGF and TNFα. Mechanistically, Atox1-depleted ECs demonstrate that Cu chaperone function of Atox1 mediated through Cu transporter ATP7A is required for VEGF-induced angiogenesis via activation of Cu enzyme lysyl oxidase. Moreover, Atox1 functions as a Cu-dependent transcription factor for NADPH oxidase organizer p47phox, thereby increasing ROS-NFκB-VCAM-1/ICAM-1 expression and monocyte adhesion in ECs inflamed with TNFα in an ATP7A-independent manner. These findings demonstrate a novel linkage between Atox1 and NADPH oxidase involved in inflammatory neovascularization and suggest Atox1 as a potential therapeutic target for treatment of ischemic disease.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.