Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
3.
Hypertension ; 76(3): 1006-1018, 2020 09.
Article in English | MEDLINE | ID: mdl-32755412

ABSTRACT

Previously, we showed that peripheral administration of 6ß-hydroxytestosterone, a CYP1B1 (cytochrome P450 1B1)-generated metabolite of testosterone, promotes angiotensin II-induced hypertension in male mice. However, the site of action and the underlying mechanism by which 6ß-hydroxytestosterone contributes to angiotensin II-induced hypertension is not known. Angiotensin II increases blood pressure by its central action, and CYP1B1 is expressed in the brain. This study was conducted to determine whether testosterone-CYP1B1 generated metabolite 6ß-hydroxytestosterone locally in the brain promotes the effect of systemic angiotensin II to produce hypertension in male mice. Central CYP1B1 knockdown in wild-type (Cyp1b1+/+) mice by intracerebroventricular-adenovirus-GFP (green fluorescence protein)-CYP1B1-short hairpin (sh)RNA attenuated, whereas reconstitution of CYP1B1 by adenovirus-GFP-CYP1B1-DNA in the paraventricular nucleus but not in subfornical organ in Cyp1b1-/- mice restored angiotensin II-induced increase in systolic blood pressure measured by tail-cuff. Intracerebroventricular-testosterone in orchidectomized (Orchi)-Cyp1b1+/+ but not in Orchi-Cyp1b1-/-, and intracerebroventricular-6ß-hydroxytestosterone in the Orchi-Cyp1b1-/- mice restored the angiotensin II-induced: (1) increase in mean arterial pressure measured by radiotelemetry, and autonomic imbalance; (2) reactive oxygen species production in the subfornical organ and paraventricular nucleus; (3) activation of microglia and astrocyte, and neuroinflammation in the paraventricular nucleus. The effect of intracerebroventricular-6ß-hydroxytestosterone to restore the angiotensin II-induced increase in mean arterial pressure and autonomic imbalance in Orchi-Cyp1b1-/- mice was inhibited by intracerebroventricular-small interfering (si)RNA-androgen receptor (AR) and GPRC6A (G protein-coupled receptor C6A). These data suggest that testosterone-CYP1B1-generated metabolite 6ß-hydroxytestosterone, most likely in the paraventricular nucleus via AR and GPRC6A, contributes to angiotensin II-induced hypertension and neuroinflammation in male mice.


Subject(s)
Cytochrome P-450 CYP1B1 , Hydroxytestosterones/metabolism , Hypertension/metabolism , Neurogenic Inflammation/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Androgen/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin II/metabolism , Animals , Blood Pressure/physiology , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Hypertension/etiology , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism
4.
Hypertension ; 75(4): 1054-1062, 2020 04.
Article in English | MEDLINE | ID: mdl-32148125

ABSTRACT

Previously, we showed that peripheral administration of 2-ME (2-methoxyestradiol), a CYP1B1 (cytochrome P450 1B1)-catechol-O-methyltransferase (COMT) generated metabolite of E2 (17ß-Estradiol), protects against angiotensin II-induced hypertension in female mice. The demonstration that central E2 inhibits angiotensin II-induced hypertension, together with the expression of CYP1B1 in the brain, led us to hypothesize that E2-CYP1B1 generated metabolite 2-ME in the brain mediates its protective action against angiotensin II-induced hypertension in female mice. To test this hypothesis, we examined the effect of intracerebroventricularly (ICV) administered E2 in ovariectomized (OVX)-wild-type (Cyp1b1+/+) and OVX-Cyp1b1-/- mice on the action of systemic angiotensin II. ICV-E2 attenuated the angiotensin II-induced increase in mean arterial blood pressure, impairment of baroreflex sensitivity, and sympathetic activity in OVX-Cyp1b1+/+ but not in ICV-injected short interfering (si)RNA-COMT or OVX-Cyp1b1-/- mice. ICV-2-ME attenuated the angiotensin II-induced increase in blood pressure in OVX-Cyp1b1-/- mice; this effect was inhibited by ICV-siRNA estrogen receptor-α (ERα) and G protein-coupled estrogen receptor 1 (GPER1). ICV-E2 in OVX-Cyp1b1+/+ but not in OVX-Cyp1b1-/- mice and 2-ME in the OVX-Cyp1b1-/- inhibited angiotensin II-induced increase in reactive oxygen species production in the subfornical organ and paraventricular nucleus, activation of microglia and astrocyte, and neuroinflammation in paraventricular nucleus. Furthermore, central CYP1B1 gene disruption in Cyp1b1+/+ mice by ICV-adenovirus-GFP (green fluorescence protein)-CYP1B1-short hairpin (sh)RNA elevated, while reconstitution by adenovirus-GFP-CYP1B1-DNA in the paraventricular nucleus but not in subfornical organ in Cyp1b1-/- mice attenuated the angiotensin II-induced increase in systolic blood pressure. These data suggest that E2-CYP1B1-COMT generated metabolite 2-ME, most likely in the paraventricular nucleus via estrogen receptor-α and GPER1, protects against angiotensin II-induced hypertension and neuroinflammation in female mice.


Subject(s)
2-Methoxyestradiol/therapeutic use , Angiotensin II/pharmacology , Blood Pressure/drug effects , Cytochrome P-450 CYP1B1/metabolism , Estradiol/pharmacology , Hypertension/prevention & control , Inflammation/prevention & control , Neuroprotective Agents/therapeutic use , 2-Methoxyestradiol/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Baroreflex/drug effects , Brain/drug effects , Brain/metabolism , Cytochrome P-450 CYP1B1/genetics , Female , Hypertension/chemically induced , Hypertension/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Mice , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
5.
J Am Heart Assoc ; 7(18): e010065, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30371217

ABSTRACT

Background We have reported that cytochrome P450 1B1 ( CYP 1B1), expressed in cardiovascular tissues, contributes to angiotensin II -induced vascular smooth muscle cell ( VSMC ) migration and proliferation and development of hypertension in various experimental animal models via generation of reactive oxygen species. This study was conducted to determine the contribution of CYP 1B1 to platelet-derived growth factor-BB-induced VSMC migration and proliferation in vitro and to neointimal growth in vivo. Methods and Results VSMC s isolated from aortas of male Cyp1b1 +/+ and Cyp1b1 -/- mice were used for in vitro experiments. Moreover, carotid arteries of Cyp1b1 +/+ and Cyp1b1 -/- mice were injured with a metal wire to assess neointimal growth after 14 days. Platelet-derived growth factor- BB -induced migration and proliferation and H2O2 production were found to be attenuated in VSMC s from Cyp1b1 -/- mice and in VSMC s of Cyp1b1 +/+ mice treated with 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl, a superoxide dismutase and catalase mimetic. In addition, wire injury resulted in neointimal growth, as indicated by increased intimal area, intima/media ratio, and percentage area of restenosis, as well as elastin disorganization and adventitial collagen deposition in carotid arteries of Cyp1b1 +/+ mice, which were minimized in Cyp1b1 -/- mice. Wire injury also increased infiltration of inflammatory and immune cells, as indicated by expression of CD 68+ macrophages and CD 3+ T cells, respectively, in the injured arteries of Cyp1b1 +/+ mice, but not Cyp1b1 -/- mice. Administration of 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl attenuated neointimal growth in wire-injured carotid arteries of Cyp1b1 +/+ mice. Conclusions These data suggest that CYP 1B1-dependent oxidative stress contributes to the neointimal growth caused by wire injury of carotid arteries of male mice.


Subject(s)
Carotid Arteries/metabolism , Carotid Artery Injuries/genetics , Cytochrome P-450 CYP1B1/genetics , Gene Expression Regulation , Neointima/metabolism , Oxidative Stress , Animals , Blotting, Western , Carotid Arteries/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP1B1/biosynthesis , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Neointima/pathology , RNA/genetics
6.
Am J Hypertens ; 31(5): 622-629, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29342227

ABSTRACT

BACKGROUND: Recently, we reported that angiotensin II (Ang II)-induced hypertension is mediated by group IV cytosolic phospholipase A2α (cPLA2α) via production of prohypertensive eicosanoids. Since Ang II increases blood pressure (BP) via its action in the subfornical organ (SFO), it led us to investigate the expression and possible contribution of cPLA2α to oxidative stress and development of hypertension in this brain area. METHODS: Adenovirus (Ad)-green fluorescence protein (GFP) cPLA2α short hairpin (sh) RNA (Ad-cPLA2α shRNA) and its control Ad-scrambled shRNA (Ad-Scr shRNA) or Ad-enhanced cyan fluorescence protein cPLA2α DNA (Ad-cPLA2α DNA) and its control Ad-GFP DNA were transduced into SFO of cPLA2α+/+ and cPLA2α-/- male mice, respectively. Ang II (700 ng/kg/min) was infused for 14 days in these mice, and BP was measured by tail-cuff and radio telemetry. cPLA2 activity, reactive oxygen species production, and endoplasmic reticulum stress were measured in the SFO. RESULTS: Transduction of SFO with Ad-cPLA2α shRNA, but not Ad-Scr shRNA in cPLA2α+/+ mice, minimized expression of cPLA2α, Ang II-induced cPLA2α activity and oxidative stress in the SFO, BP, and cardiac and renal fibrosis. In contrast, Ad-cPLA2α DNA, but not its control Ad-GFP DNA in cPLA2α-/- mice, restored the expression of cPLA2α, and Ang II-induced increase in cPLA2 activity and oxidative stress in the SFO, BP, cardiac, and renal fibrosis. CONCLUSIONS: These data suggest that cPLA2α in the SFO is crucial in mediating Ang II-induced hypertension and associated pathogenesis. Therefore, development of selective cPLA2α inhibitors could be useful in treating hypertension and its pathogenesis.


Subject(s)
Angiotensin II/pharmacology , Brain/enzymology , Group IV Phospholipases A2/physiology , Hypertension/etiology , Reactive Oxygen Species/metabolism , Animals , Collagen/metabolism , Endoplasmic Reticulum Stress , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/genetics , Hypertension/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
Am J Pathol ; 186(8): 2204-2219, 2016 08.
Article in English | MEDLINE | ID: mdl-27301358

ABSTRACT

Cytochrome P450 (CYP) 1B1 is implicated in vascular smooth muscle cell migration, proliferation, and hypertension. We assessed the contribution of CYP1B1 to angiotensin (Ang) II-induced abdominal aortic aneurysm (AAA). Male Apoe(-/-)/Cyp1b1(+/+) and Apoe(-/-)/Cyp1b1(-/-) mice were infused with Ang II or its vehicle for 4 weeks; another group of Apoe(-/-)/Cyp1b1(+/+) mice was coadministered the CYP1B1 inhibitor 2,3',4,5'-tetramethoxystilbene (TMS) every third day for 4 weeks. On day 28 of Ang II infusion, AAAs were analyzed by ultrasound and ex vivo by Vernier calipers, mice were euthanized, and tissues were harvested. Ang II produced AAAs in Apoe(-/-)/Cyp1b1(+/+) mice; mice treated with TMS or Apoe(-/-)/Cyp1b1(-/-) mice had reduced AAAs. Ang II enhanced infiltration of macrophages, T cells, and platelets and increased platelet-derived growth factor D, Pdgfrb, Itga2, and matrix metalloproteinases 2 and 9 expression in aortic lesions; these changes were inhibited in mice treated with TMS and in Apoe(-/-)/Cyp1b1(-/-) mice. Oxidative stress resulted in cyclooxygenase-2 expression in aortic lesions. These effects were minimized in Apoe(-/-)/Cyp1b1(+/+) mice treated with TMS and in Apoe(-/-)/Cyp1b1(-/-) mice and by concurrent treatment with the superoxide scavenger 4-hydroxyl-2,2,6,6-tetramethylpiperidine-1-oxyl. CYP1B1 contributed to the development of Ang II-induced AAA and associated pathogenic events in mice, likely by enhancing oxidative stress and associated signaling events. Thus, CYP1B1 may serve as a target for therapeutic agents for AAA in males.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Cytochrome P-450 CYP1B1/metabolism , Oxidative Stress/physiology , Angiotensin II/toxicity , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
8.
Hypertension ; 67(1): 206-13, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26573711

ABSTRACT

Cytochrome P450 (CYP) 1B1 contributes to vascular smooth muscle cell growth and hypertension in male mice. This study was conducted to determine the contribution of CYP1B1 to the development of atherosclerosis and hypertension and associated pathogenesis in 8-week-old male apolipoprotein E-deficient (ApoE(-/-)/Cyp1b1(+/+)), and ApoE- and CYP1B1-deficient (ApoE(-/-)/Cyp1b1(-/-)) mice fed a normal or atherogenic diet for 12 weeks. A separate group of ApoE(-/-)/Cyp1b1(+/+) mice on an atherogenic diet was injected every third day with the CYP1B1 inhibitor, 2,3',4,5'-tetramethoxystilbene (300 µg/kg), or its vehicle, dimethyl sulfoxide (30 µL, IP); systolic blood pressure was measured by the tail cuff method. After 12 weeks, mice were euthanized, blood collected for lipid analysis, and aortas harvested for measuring lesions and remodeling, and for infiltration of inflammatory cells by histological and immunohistochemical analysis, respectively, and for reactive oxygen species production. Blood pressure, areas of lipids and collagen deposition, elastin breaks, infiltration of macrophages and T lymphocytes, reactive oxygen species generation in the aorta, and plasma lipid levels were increased in ApoE(-/-)/Cyp1b1(+/+) mice on an atherogenic diet; these changes were minimized in mice given 2,3',4,5'-tetramethoxystilbene, and in ApoE(-/-)/Cyp1b1(-/-) mice on an atherogenic diet; absorption/production of lipids remained unaltered in these mice. These data suggest that aortic lesions, hypertension, and associated pathogenesis in ApoE(-/-)/Cyp1b1(+/+) mice on an atherogenic diet are most likely dependent on CYP1B1-generated oxidative stress and increased plasma lipid levels independent of blood pressure and absorption of lipids. CYP1B1 could serve as a novel target for developing drugs to treat atherosclerosis and hypertension caused by hypercholesterolemia.


Subject(s)
Atherosclerosis/genetics , Blood Pressure/physiology , Cytochrome P-450 CYP1B1/genetics , Endothelium, Vascular/metabolism , Gene Expression Regulation , Hypertension/genetics , RNA/genetics , Animals , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Cytochrome P-450 CYP1B1/biosynthesis , Disease Models, Animal , Endothelium, Vascular/physiopathology , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice , Mice, Knockout , Vasodilation
9.
Am J Hypertens ; 29(2): 258-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26045535

ABSTRACT

BACKGROUND: The kidney plays an important role in regulating blood pressure (BP). cPLA2α in the kidney is activated by various agents including angiotensin II (Ang II) and selectively releases arachidonic acid (AA) from tissue lipids, generating pro- and antihypertensive eicosanoids. Since activation of cPLA2α is the rate-limiting step in AA release, this study was conducted to determine its contribution to renal dysfunction and end-organ damage associated with Ang II-induced hypertension. METHODS: cPLA2α(+/+) and cPLA2α(-/-) mice were infused with Ang II (700 ng/ kg/min) or its vehicle for 13 days. Mice were placed in metabolic cages to monitor their food and water intake, and urine was collected and its volume was measured. Doppler imaging was performed to assess renal hemodynamics. On the 13th day of Ang II infusion, mice were sacrificed and their tissues and blood collected for further analysis. RESULTS: Ang II increased renal vascular resistance, water intake, and urine output and Na(+) excretion, decreased urine osmolality, and produced proteinuria in cPLA2α(+/+) mice. Ang II also caused accumulation of F4/80(+) macrophages and CD3(+) T cells and renal fibrosis, and increased oxidative stress in the kidneys of cPLA2α(+/+) mice. All these effects of Ang II were minimized in cPLA2α(-/-) mice. CONCLUSION: cPLA2α contributes to renal dysfunction, inflammation, and end-organ damage, most likely via the action of pro-hypertensive eicosanoids and increased oxidative stress associated with Ang II-induced hypertension. Thus, cPLA2α could serve as a potential therapeutic target for treating renal dysfunction and end-organ damage in hypertension.


Subject(s)
Group IV Phospholipases A2/metabolism , Hypertension/enzymology , Kidney Diseases/etiology , Angiotensin II , Animals , Endothelins/blood , Fibrosis , Group IV Phospholipases A2/genetics , Hypertension/complications , Kidney/metabolism , Kidney/pathology , Kidney Diseases/pathology , Male , Mice , Renal Circulation , Vascular Resistance
10.
Hypertension ; 65(4): 784-92, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25667212

ABSTRACT

Angiotensin II activates cytosolic phospholipase A(2)α (cPLA2α) and releases arachidonic acid from tissue phospholipids, which mediate or modulate ≥1 cardiovascular effects of angiotensin II and has been implicated in hypertension. Because arachidonic acid release is the rate limiting step in eicosanoid production, cPLA2α might play a central role in the development of angiotensin II-induced hypertension. To test this hypothesis, we investigated the effect of angiotensin II infusion for 13 days by micro-osmotic pumps on systolic blood pressure and associated pathogenesis in wild type (cPLA2α(+/+)) and cPLA2α(-/-) mice. Angiotensin II-induced increase in systolic blood pressure in cPLA2α(+/+) mice was abolished in cPLA2α(-/-) mice; increased systolic blood pressure was also abolished by the arachidonic acid metabolism inhibitor, 5,8,11,14-eicosatetraynoic acid in cPLA2α(+/+) mice. Angiotensin II in cPLA2α(+/+) mice increased cardiac cPLA2 activity and urinary eicosanoid excretion, decreased cardiac output, caused cardiovascular remodeling with endothelial dysfunction, and increased vascular reactivity in cPLA2α(+/+) mice; these changes were diminished in cPLA2α(-/-) mice. Angiotensin II also increased cardiac infiltration of F4/80(+) macrophages and CD3(+) T lymphocytes, cardiovascular oxidative stress, expression of endoplasmic reticulum stress markers p58(IPK), and CHOP in cPLA2α(+/+) but not cPLA2α(-/-) mice. Angiotensin II increased cardiac activity of ERK1/2 and cSrc in cPLA2α(+/+) but not cPLA2α(-/-) mice. These data suggest that angiotensin II-induced hypertension and associated cardiovascular pathophysiological changes are mediated by cPLA2α activation, most likely through the release of arachidonic acid and generation of eicosanoids with predominant prohypertensive effects and activation of ≥1 signaling molecules, including ERK1/2 and cSrc.


Subject(s)
Blood Pressure/physiology , Cytosol/enzymology , Gene Expression Regulation , Group IV Phospholipases A2/genetics , Hypertension/genetics , Oxidative Stress , RNA/genetics , Angiotensin II/toxicity , Animals , Disease Models, Animal , Group IV Phospholipases A2/biosynthesis , Hypertension/chemically induced , Hypertension/metabolism , Lipid Metabolism , Mice , Mice, Inbred BALB C , Myocardium/enzymology , Myocardium/pathology , Real-Time Polymerase Chain Reaction
11.
J Nanosci Nanotechnol ; 13(9): 6312-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24205651

ABSTRACT

In this research, we will present Al doped ZnO thin films for transparent conducting oxide applications. Aluminum doped zinc oxide (AZO) thin films have been deposited on the glass substrates by sol-gel spin-coating method using zinc acetate dehydrate (Zn(CH3COO)2 2H2O) and aluminum chloride hexahydrate (AlCl3 x 6H2O) as cation sources. In this study, we investigated the effects of near infrared ray (NIR) annealing on the structural, optical and electrical characteristics of the AZO thin films. The experimental results showed that AZO thin films have a hexagonal wurtzite crystal structure and had a good transmittance higher than 85% within the visible wavelength region. It was also found that the additional energy of NIR helps to improve the electrical properties of Al doped ZnO transparent conducting oxides.

12.
Prostaglandins Other Lipid Mediat ; 98(3-4): 69-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22210049

ABSTRACT

The aim of this review is to discuss the contribution of cytochrome P450 (CYP) 1B1 in vascular smooth muscle cell growth, hypertension, and associated pathophysiology. CYP1B1 is expressed in cardiovascular and renal tissues, and mediates angiotensin II (Ang II)-induced activation of NADPH oxidase and generation of reactive oxygen species (ROS), and vascular smooth muscle cell migration, proliferation, and hypertrophy. Moreover, CYP1B1 contributes to the development and/or maintenance of hypertension produced by Ang II-, deoxycorticosterone (DOCA)-salt-, and N(ω)-nitro-L-arginine methyl ester-induced hypertension and in spontaneously hypertensive rats. The pathophysiological changes, including cardiovascular hypertrophy, increased vascular reactivity, endothelial and renal dysfunction, injury and inflammation associated with Ang II- and/or DOCA-salt induced hypertension in rats, and Ang II-induced hypertension in mice are minimized by inhibition of CYP1B1 activity with 2,4,3',5'-tetramethoxystilbene or by Cyp1b1 gene disruption in mice. These pathophysiological changes appear to be mediated by increased production of ROS via CYP1B1-dependent NADPH oxidase activity and extracellular signal-regulated kinase 1/2, p38 mitogen-activated protein kinase, and c-Src.


Subject(s)
Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Hypertension/drug therapy , Hypertension/physiopathology , Animals , Cardiovascular System/enzymology , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Cell Movement/drug effects , Humans , Hypertension/enzymology , Hypertension/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology
13.
Am J Physiol Renal Physiol ; 302(4): F408-20, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22088434

ABSTRACT

We investigated the contribution of cytochrome P-450 1B1 (CYP1B1) to renal dysfunction and organ damage associated with ANG II-induced hypertension in rats. ANG II (300 ng·kg(-1)·min(-1)) or vehicle were infused for 2 wk, with daily injections of a selective CYP1B1 inhibitor, 2,4,3',5'-tetramethoxystilbene (TMS; 300 µg/kg ip), or its vehicle. ANG II increased blood pressure and renal CYP1B1 activity that were prevented by TMS. ANG II also increased water intake and urine output, decreased glomerular filtration rate, increased urinary Na(+) and K(+) excretion, and caused proteinuria, all of which were prevented by TMS. ANG II infusion caused hypertrophy, endothelial dysfunction, and increased reactivity of renal and interlobar arteries to vasoconstrictor agents and renal vascular resistance and interstitial fibrosis as indicated by accumulation of α-smooth muscle actin, fibronectin, and collagen, and inflammation as indicated by increased infiltration of CD-3(+) cells; these effects were inhibited by TMS. ANG II infusion also increased production of reactive oxygen species (ROS) and activities of NADPH oxidase, ERK1/2, p38 MAPK, and c-Src that were prevented by TMS. TMS alone had no effect on any of the above parameters. These data suggest that CYP1B1 contributes to the renal pathophysiological changes associated with ANG II-induced hypertension, most likely via increased ROS production and activation of ERK1/2, p38 MAPK, and c-Src and that CYP1B1 could serve as a novel target for treating renal disease associated with hypertension.


Subject(s)
Angiotensin II/toxicity , Aryl Hydrocarbon Hydroxylases/metabolism , Hypertension, Renal/enzymology , Kidney/enzymology , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Blood Pressure/drug effects , Blood Pressure/physiology , Cytochrome P-450 CYP1B1 , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hemodynamics/drug effects , Hemodynamics/physiology , Hypertension, Renal/chemically induced , Hypertension, Renal/physiopathology , Inflammation/enzymology , Inflammation/physiopathology , Kidney/blood supply , Kidney/drug effects , Kidney/physiopathology , Male , NADPH Oxidases/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Stilbenes/pharmacology
14.
Am J Physiol Heart Circ Physiol ; 301(3): H1043-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642504

ABSTRACT

Spleen tyrosine kinase (Syk), expressed in endothelial cells, has been implicated in migration and proliferation and in vasculogenesis. This study was conducted to determine the contribution of Syk and the underlying mechanism to the angiogenic effect of ANG II and VEGF. Angiogenesis was determined by tube formation from the endothelial cell line EA.hy926 (EA) and human umbilical vein endothelial cells (HUVECs) and microvessel sprouting in rat aortic rings. ANG II (10 nM), EGF (30 ng/ml), and VEGF (50 ng/ml) stimulated EA cells and HUVECs to form tubular networks and increased aortic sprouting; these effects were blocked by VEGF receptor-1 and Flt-1 antibody (Flt-1/Fc) but not by the VEGF receptor-2 (Flk-1) antagonist SU-1498. ANG II increased the phosphorylation of Flt-1 but not Flk-1, whereas VEGF increased the phosphorylation of both receptors in EA cells and HUVECs. VEGF expression elicited by ANG II was not altered by Flt-1/Fc or SU-1498. EGF stimulated tube formation from EA cells and HUVECs and Flt-1 phosphorylation and aortic sprouting, which were blocked by the EGF receptor antagonist AG-1478 and Flt-1/Fc but not by SU-1498. ANG II-, EGF-, and VEGF-induced tube formation and aortic sprouting were attenuated by the Syk inhibitor piceatannol and by Syk short hairpin interfering (sh)RNA and small interfering RNA, respectively. ANG II, EGF, and VEGF increased Syk phosphorylation, which was inhibited by piceatannol and Syk shRNA in EA cells and HUVECs. Neither piceatannol nor Syk shRNA altered ANG II-, EGF-, or VEGF-induced phosphorylation of Flt-1. These data suggest that ANG II stimulates angiogenesis via transactivation of the EGF receptor, which promotes the phosphorylation of Flt-1 and activation of Syk independent of VEGF expression.


Subject(s)
Angiotensin II/metabolism , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neovascularization, Physiologic , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism , Analysis of Variance , Angiogenesis Inhibitors/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Activation , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Neovascularization, Physiologic/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , RNA Interference , Rats , Rats, Sprague-Dawley , Signal Transduction , Syk Kinase , Time Factors , Transfection , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Am J Physiol Heart Circ Physiol ; 299(6): H1891-901, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20852048

ABSTRACT

Reactive oxygen species (ROS) contribute to various models of hypertension, including deoxycorticosterone acetate (DOCA)-salt-induced hypertension. Recently, we have shown that ROS, generated by cytochrome P-450 1B1 (CYP1B1) from arachidonic acid, mediate vascular smooth muscle cell growth caused by angiotensin II. This study was conducted to determine the contribution of CYP1B1 to hypertension and associated pathophysiological changes produced by DOCA (30 mg/kg) given subcutaneously per week with 1% NaCl + 0.1% KCl in drinking water to uninephrectomized rats for 6 wk. DOCA-salt treatment increased systolic blood pressure (SBP). Injections of the selective inhibitor of CYP1B1, 2,3',4,5'-tetramethoxystilbene (TMS; 300 µg/kg ip every 3rd day) initiated at the 4th week of DOCA-salt treatment normalized SBP and decreased CYP1B1 activity but not its expression in the aorta, heart, and kidney. TMS also inhibited cardiovascular and kidney hypertrophy, prevented the increase in vascular reactivity and endothelial dysfunction, and minimized the increase in urinary protein and K(+) output and the decrease in urine osmolality, Na(+) output, and creatinine clearance associated with DOCA-salt treatment. These pathophysiological changes caused by DOCA-salt treatment and associated increase in vascular superoxide production, NADPH oxidase activity, and expression of NOX-1, and ERK1/2 and p38 MAPK activities in the aorta, heart, and kidney were inhibited by TMS. These data suggest that CYP1B1 contributes to DOCA-salt-induced hypertension and associated pathophysiological changes, most likely as a result of increased ROS production and ERK1/2 and p38 MAPK activity, and could serve as a novel target for the development of agents like TMS to treat hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Blood Pressure/drug effects , Desoxycorticosterone , Enzyme Inhibitors/pharmacology , Hypertension/prevention & control , Sodium Chloride, Dietary , Stilbenes/pharmacology , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/blood , Animals , Aorta/drug effects , Aorta/enzymology , Aryl Hydrocarbon Hydroxylases/metabolism , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Cytochrome P-450 CYP1B1 , Disease Models, Animal , Diuresis/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Hydroxyeicosatetraenoic Acids/blood , Hypertension/enzymology , Hypertension/etiology , Hypertension/pathology , Hypertension/physiopathology , Kidney/drug effects , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Myocardium/enzymology , Myocardium/pathology , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Proteinuria/enzymology , Proteinuria/physiopathology , Proteinuria/prevention & control , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , Time Factors , Vasoconstriction/drug effects , Vasodilation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Hypertension ; 55(6): 1461-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20439821

ABSTRACT

Cytochrome P450 1B1, expressed in vascular smooth muscle cells, can metabolize arachidonic acid in vitro into several products including 12- and 20-hydroxyeicosatetraenoic acids that stimulate vascular smooth muscle cell growth. This study was conducted to determine whether cytochrome P450 1B1 contributes to angiotensin II-induced rat aortic smooth muscle cell migration, proliferation, and protein synthesis. Angiotensin II stimulated migration of these cells, measured by the wound healing approach, by 1.78-fold; and DNA synthesis, measured by [(3)H]thymidine incorporation, by 1.44-fold after 24 hours; and protein synthesis, measured by [(3)H]leucine incorporation, by 1.40-fold after 48 hours. Treatment of vascular smooth muscle cells with the cytochrome P450 1B1 inhibitor 2,4,3',5'-tetramethoxystilbene or transduction of these cells with adenovirus cytochrome P450 1B1 small hairpin RNA but not its scrambled control reduced the activity of this enzyme and abolished angiotensin II- and arachidonic acid-induced cell migration, as well as [(3)H]thymidine and [(3)H]leucine incorporation. Metabolism of arachidonic acid to 5-, 12-, 15-, and 20-hydoxyeicosatetraenoic acids in these cells was not altered, but angiotensin II- and arachidonic acid-induced reactive oxygen species production and extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase activity were inhibited by 2,4,3',5'-tetramethoxystilbene and cytochrome P450 1B1 small hairpin RNA (shRNA) and by Tempol, which inactivates reactive oxygen species. Tempol did not alter cytochrome P450 1B1 activity. These data suggest that angiotensin II-induced vascular smooth muscle cell migration and growth are mediated by reactive oxygen species generated from arachidonic acid by cytochrome P450 1B1 and activation of extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase.


Subject(s)
Angiotensin II/pharmacology , Cell Movement/drug effects , Cytochrome P-450 CYP2B1/metabolism , Muscle, Smooth, Vascular/drug effects , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Animals , Aorta/cytology , Cell Movement/physiology , Cell Proliferation/drug effects , Cells, Cultured , Cytochrome P-450 CYP2B1/drug effects , DNA/metabolism , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/physiology , Proteins/metabolism , Rats , Renin-Angiotensin System/physiology , Sensitivity and Specificity
17.
Kidney Blood Press Res ; 33(1): 24-9, 2010.
Article in English | MEDLINE | ID: mdl-20185928

ABSTRACT

Injured podocytes proliferate in cellular focal segmental glomerulosclerosis (FSGS), collapsing FSGS and crescentic glomerulonephritis, where TGF-beta(1) is overexpressed in hyperplastic podocytes. Yet effects of podocyte TGF-beta on podocyte growth and development of glomerulosclerosis have not been clearly defined. TGF-beta activates Smads, Ras/extracellular signal-regulated kinase (ERK) and phosphatidyl inositol-3-kinase (PI3K) pathways in podocytes, of which the major TGF-beta/Smad signaling pathway appears to override the minor TGF-beta-induced Ras/ERK/PI3K pathways. We provide evidence that increasedTGF-beta/Smad signaling activity by hyperplastic podocytes may lead to mesangial cell matrix overproduction and eventually to podocyte apoptosis and/or detachment, culminating in the development of glomerulosclerosis. In this regard, TGF-beta, which is overexpressed by hyperplastic podocytes, may play an important role for the cellular and collapsing variants of FSGS to evolve into the classic FSGS pattern. In contrast, podocyte proliferation that is induced by Ras/ERK signaling activity in proliferative podocyte diseases seems to be mostly independent of TGF-beta(1) activity. Collectively, these data bring new insights into our understanding of the overexpression of TGF-beta in hyperplastic podocytes in progressive glomerular diseases.


Subject(s)
Glomerulosclerosis, Focal Segmental/metabolism , Podocytes/pathology , Podocytes/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans
18.
J Pharmacol Exp Ther ; 332(1): 116-24, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19797620

ABSTRACT

Angiotensin II (Ang II) stimulates protein synthesis by activating spleen tyrosine kinase (Syk) and DNA synthesis through epidermal growth factor receptor (EGFR) transactivation in vascular smooth muscle cells (VSMCs). This study was conducted to determine whether Syk mediates Ang II-induced migration of aortic VSMCs using a scratch wound approach. Treatment with Ang II (200 nM) for 24 h increased VSMC migration by 1.56 +/- 0.14-fold. Ang II-induced VSMC migration and Syk phosphorylation as determined by Western blot analysis were minimized by the Syk inhibitor piceatannol (10 microM) and by transfecting VSMCs with dominant-negative but not wild-type Syk plasmid. Ang II-induced VSMC migration and Syk phosphorylation were attenuated by inhibitors of c-Src [4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2)], p38 mitogen-activated protein kinase (MAPK) [4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)1H-imidazole (SB202190)], and extracellular signal-regulated kinase (ERK) 1/2 [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio) butadiene (U0126)]. SB202190 attenuated p38 MAPK and c-Src but not ERK1/2 phosphorylation, indicating that p38 MAPK acts upstream of c-Src and Syk. The c-Src inhibitor PP2 attenuated Syk and ERK1/2 phosphorylation, suggesting that c-Src acts upstream of Syk and ERK1/2. Ang II- and epidermal growth factor (EGF)-induced VSMC migration and EGFR phosphorylation were inhibited by the EGFR blocker 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) (2 microM). Neither the Syk inhibitor piceatannol nor the dominant-negative Syk mutant altered EGF-induced cell migration or Ang II- and EGF-induced EGFR phosphorylation. The c-Src inhibitor PP2 diminished EGF-induced VSMC migration and EGFR, ERK1/2, and p38 MAPK phosphorylation. The ERK1/2 inhibitor U0126 (10 microM) attenuated EGF-induced cell migration and ERK1/2 but not EGFR phosphorylation. These data suggest that Ang II stimulates VSMC migration via p38 MAPK-activated c-Src through Syk and via EGFR transactivation through ERK1/2 and partly through p38 MAPK.


Subject(s)
Angiotensin II/pharmacology , Cell Movement/drug effects , Epidermal Growth Factor/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Protein-Tyrosine Kinases/metabolism , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases/metabolism , Angiotensin II/physiology , Animals , Aorta, Thoracic/cytology , Butadienes/pharmacology , CSK Tyrosine-Protein Kinase , Cell Culture Techniques , Cell Migration Assays , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Nitriles/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines , Rats , Rats, Sprague-Dawley , Syk Kinase , Transcriptional Activation/drug effects , Tyrphostins/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , src-Family Kinases
19.
Histol Histopathol ; 24(7): 901-8, 2009 07.
Article in English | MEDLINE | ID: mdl-19475536

ABSTRACT

Glomerulosclerosis is characterized by mesangial matrix accumulation that is mediated primarily by activation of transforming growth factor-beta (TGF-beta). Unlike podocytes, mesangial cells secrete TGF-beta in response to common in vitro fibrogenic stimuli. However, mesangial immunostaining for active TGF-beta1 in chronic glomerular disease is almost negligible, despite increased mesangial TGF-beta1 mRNA expression, while podocytes covering the sclerotic glomerular segments exhibit increased TGF-beta1 protein expression. The mechanisms whereby TGF-beta is activated in the diseased glomeruli and how the activated TGF-beta leads to mesangial matrix overproduction are not clear. We provide evidence that TGF-beta secreted as latent complexes by mesangial cells is stored in the mesangial matrix, from which soluble forms of latent TGF-beta are released and localized to the podocyte surface in chronic glomerular disease. Podocyte-derived reactive oxygen species, plasmin and thrombospondin-1, particularly renin-angiotensin-aldosterone system-induced oxidative stress, seem to be involved in TGF-beta activation in podocytes. We also provide evidence that the TGF-beta-induced secretion of connective tissue growth factor and vascular endothelial growth factor by podocytes acts as a paracrine regulatory mechanism on mesangial cells, which may cause mesangial matrix accumulation culminating in the development of glomerulosclerosis. Collectively, these data bring new insights into our understanding of the roles of the mesangial cells and podocytes in the TGF-beta-induced mesangial matrix synthesis in chronic glomerular disease.


Subject(s)
Glomerular Mesangium/metabolism , Kidney Glomerulus/pathology , Podocytes/metabolism , Sclerosis/pathology , Transforming Growth Factor beta/metabolism , Animals , Chronic Disease , Extracellular Matrix/metabolism , Glomerular Mesangium/cytology , Humans , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
20.
Am J Nephrol ; 29(1): 62-70, 2009.
Article in English | MEDLINE | ID: mdl-18689980

ABSTRACT

Glomerulosclerosis frequently complicates most renal diseases and is characterized by mesangial matrix accumulation. Oxidized low-density lipoprotein (Ox-LDL) could induce oxidative stress and profibrotic gene expression in mesangial cells. This article will review our current understanding of the pathogenetic mechanisms of lipid-mediated glomerulosclerosis, emphasizing the fibrogenic signaling cascades triggered by Ox-LDL. In addition, therapeutic strategies to prevent the development of Ox-LDL-mediated glomerulosclerosis will be discussed.


Subject(s)
Glomerulonephritis/metabolism , Lipoproteins, LDL/metabolism , Oxidative Stress , Antioxidants/metabolism , Enzyme Activation , Glomerulonephritis/pathology , Humans , Lipid Peroxidation , MAP Kinase Signaling System , Mesangial Cells/metabolism , Mesangial Cells/pathology , NADPH Oxidases/metabolism , Oxygen/metabolism , Protein Kinase C/antagonists & inhibitors , Renin-Angiotensin System , Signal Transduction , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...