Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 12(11)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-38001814

ABSTRACT

BACKGROUND AND AIMS: we previously reported in studies on organoid-cultured bovine pulmonary arteries that pulmonary hypertension (PH) conditions of exposure to hypoxia or endothelin-1 caused a loss of a cartilage oligomeric matrix protein (COMP) stabilization of bone morphogenetic protein receptor-2 (BMPR2) function, a known key process contributing to pulmonary hypertension development. Based on subsequent findings, these conditions were associated with an extracellular superoxide-mediated increase in matrix metalloproteinase 9 (MMP-9) expression. We investigated if this contributed to PH development using mice deficient in MMP9. RESULTS: wild-type (WT) mice exposed to Sugen/Hypoxia (SuHx) to induce PH had increased levels of MMP9 in their lungs. Hemodynamic measures from MMP9 knockout mice (MMP9 KO) indicated they had attenuated PH parameters compared to WT mice based on an ECHO assessment of pulmonary artery pressure, right ventricular systolic pressure, and Fulton index hypertrophy measurements. In vitro vascular reactivity studies showed impaired endothelium-dependent and endothelium-independent NO-associated vasodilatory responses in the pulmonary arteries of SuHx mice and decreased lung levels of COMP and BMPR2 expression. These changes were attenuated in MMP9 KO mice potentially through preserving COMP-dependent stabilization of BMPR2. INNOVATION: this study supports a new function of superoxide in increasing MMP9 and the associated impairment of BMPR2 in promoting PH development which could be a target for future therapies. CONCLUSION: superoxide, through promoting increases in MMP9, mediates BMPR2 depletion and its consequent control of vascular function in response to PH mediators and the SuHx mouse model of PH.

2.
Am J Physiol Heart Circ Physiol ; 325(4): H645-H655, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37505471

ABSTRACT

The endothelial glycocalyx (EG) is degraded early during sepsis, and currently available treatments are not effective in promptly restoring it. Here, we created liposomal nanocarriers of preassembled glycocalyx (LNPG) by synthesizing glycosylated syndecan-1 and inserting it into the lipid membrane of unilamellar liposomes. We hypothesized that LNPG would fuse with the endothelial cells where EG is degraded and restore EG in sepsis. We induced endotoxemia in C57BL/6J mice using lipopolysaccharides (LPS) and treated them with LNPG, saline, syndecan-1, or liposomes. LNPG significantly prolonged the survival time of LPS-treated mice compared with the other treatments. Immunostaining of en face mesenteric arteries of LPS-treated mice showed that syndecan-1 was fully restored after LNPG administration. In addition, EG height in microvasculature of mouse cremaster muscle was monitored using sidestream dark field imaging. LNPG restored the perfused boundary region (PBR), which is inversely related to EG dimensions, to the control level after LPS administration. Furthermore, flow-induced dilation in isolated mouse mesenteric arterioles was fully recovered after LNPG treatment in LPS-treated mice. In summary, our findings provide evidence of the therapeutic efficacy of LNPG in the LPS-induced mouse model of sepsis, achieved by expeditiously restoring EG through fusion of LNPG with the endothelial plasma membrane and recovery of endothelial function.NEW & NOTEWORTHY Vascular endothelial cells represent the first line of exposure to bacterial endotoxins. Here, we propose a novel therapeutic strategy using liposomes to deliver preassembled glycocalyx to vascular endothelial cell surface and consequently restore endothelial glycocalyx (EG). We tested liposomal nanocarriers of preassembled glycocalyx (LNPG) in vivo and ex vivo to establish for the first time their expeditious therapeutic efficacy in improving survival of lipopolysaccharides (LPS)-treated mice, as achieved by the restoration of EG and recovery of endothelial function.


Subject(s)
Endothelial Cells , Endotoxemia , Animals , Mice , Endothelial Cells/metabolism , Lipopolysaccharides/toxicity , Glycocalyx/metabolism , Syndecan-1/metabolism , Mice, Inbred C57BL , Endotoxemia/chemically induced , Liposomes/metabolism , Liposomes/pharmacology
3.
Adv Exp Med Biol ; 1304: 147-164, 2021.
Article in English | MEDLINE | ID: mdl-34019268

ABSTRACT

Inflammatory signaling is a major component in the development and progression of many lung diseases, including asthma, chronic obstructive pulmonary disorder (COPD), and pulmonary hypertension (PH). This chapter will provide a brief overview of asthma, COPD, and PH and how inflammation plays a vital role in these diseases. Specifically, we will discuss the role of reactive oxygen species (ROS) and Ca2+ signaling in inflammatory cellular responses and how these interactive signaling pathways mediate the development of asthma, COPD, and PH. We will also deliberate the key cellular responses of pulmonary arterial (PA) smooth muscle cells (SMCs) and airway SMCs (ASMCs) in these devastating lung diseases. The analysis of the importance of inflammation will shed light on the key questions remaining in this field and highlight molecular targets that are worth exploring. The crucial findings will not only demonstrate the novel roles of essential signaling molecules such as Rieske iron-sulfur protein and ryanodine receptor in the development and progress of asthma, COPD, and PH but also offer advanced insight for creating more effective and new therapeutic targets for these devastating inflammatory lung diseases.


Subject(s)
Asthma , Hypertension, Pulmonary , Pulmonary Disease, Chronic Obstructive , Calcium Signaling , Humans , Inflammation , Reactive Oxygen Species
4.
Adv Exp Med Biol ; 1303: 107-127, 2021.
Article in English | MEDLINE | ID: mdl-33788190

ABSTRACT

In addition to studies focused on estrogen mediation of sex-different regulation of systemic circulations, there is now increasing clinical relevance and research interests in the pulmonary circulation, in terms of sex differences in the morbidity and mortality of lung diseases such as inherent-, allergic- and inflammatory-based events. Thus, female predisposition to pulmonary artery hypertension (PAH) is an inevitable topic. To better understand the nature of sexual differentiation in the pulmonary circulation, and how heritable factors, in vivo- and/or in vitro-altered estrogen circumstances and changes in the live environment work in concert to discern the sex bias, this chapter reviews pulmonary events characterized by sex-different features, concomitant with exploration of how alterations of genetic expression and estrogen metabolisms trigger the female-predominant pathological signaling. We address the following: PAH (Sect.7.2) is characterized as an estrogenic promotion of its incidence (Sect. 7.2.2), as a function of specific germline mutations, and as an estrogen-elicited protection of its prognosis (Sect.7.2.1). More detail is provided to introduce a less recognized gene of Ephx2 that encodes soluble epoxide hydrolase (sEH) to degrade epoxyeicosatrienic acids (EETs). As a susceptible target of estrogen, Ephx2/sEH expression is downregulated by an estrogen-dependent epigenetic mechanism. Increases in pulmonary EETs then evoke a potentiation of PAH generation, but mitigation of its progression, a phenomenon similar to the estrogen-paradox regulation of PAH. Additionally, the female susceptibility to chronic obstructive pulmonary diseases (Sect. 7.3) and asthma (Sect.7.4), but less preference to COVID-19 (Sect. 7.5), and roles of estrogen in their pathogeneses are briefly discussed.


Subject(s)
COVID-19 , Hypertension, Pulmonary , Lung Diseases , Estrogens , Female , Genetic Predisposition to Disease , Humans , Lung Diseases/epidemiology , Lung Diseases/genetics , Male , Prevalence , SARS-CoV-2 , Sexism
5.
Nat Commun ; 11(1): 3527, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669538

ABSTRACT

Ca2+ signaling in pulmonary arterial smooth muscle cells (PASMCs) plays an important role in pulmonary hypertension (PH). However, the underlying specific ion channel mechanisms remain largely unknown. Here, we report ryanodine receptor (RyR) channel activity and Ca2+ release both are increased, and association of RyR2 by FK506 binding protein 12.6 (FKBP12.6) is decreased in PASMCs from mice with chronic hypoxia (CH)-induced PH. Smooth muscle cell (SMC)-specific RyR2 knockout (KO) or Rieske iron-sulfur protein (RISP) knockdown inhibits the altered Ca2+ signaling, increased nuclear factor (NF)-κB/cyclin D1 activation and cell proliferation, and CH-induced PH in mice. FKBP12.6 KO or FK506 treatment enhances CH-induced PH, while S107 (a specific stabilizer of RyR2/FKBP12.6 complex) produces an opposite effect. In conclusion, CH causes RISP-dependent ROS generation and FKBP12.6/RyR2 dissociation, leading to PH. RISP inhibition, RyR2/FKBP12.6 complex stabilization and Ca2+ release blockade may be potentially beneficial for the treatment of PH.


Subject(s)
Cyclin D1/metabolism , Electron Transport Complex III/metabolism , Hypertension, Pulmonary/metabolism , NF-kappa B/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Calcium Signaling , Cell Proliferation , Cytosol/metabolism , Humans , Hypoxia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , Oxygen/metabolism , Pulmonary Artery/pathology , Reactive Oxygen Species/metabolism , Respiration Disorders/metabolism , Signal Transduction
6.
Article in English | MEDLINE | ID: mdl-32633545

ABSTRACT

20-Hydroxyeicosatetraenoic acid (20-HETE) has been linked to blood pressure (BP) regulation via actions on the renal microvasculature and tubules. We assessed tubular 20-HETE contribution to hypertension by generating transgenic mice overexpressing the CYP4A12-20-HETE synthase (PT-4a12 mice) under the control of the proximal tubule (PT)-specific promoter, phosphoenolpyruvate carboxykinase (PEPCK). 20-HETE levels in the kidney cortex of male (967±210 vs. 249±69 pg/mg protein), but not female (121±15 vs. 92±11 pg/mg protein) PT-4a12 mice, showed a 2.5-fold increase compared to WT. Renal cortical Cyp4a12 mRNA and CYP4A12 protein in male, but not female PT-4a12 mice increased by 2-3-fold compared to WT. Male PT-4a12 mice displayed elevated BP (142±1 vs. 111±4 mmHg, p<0.0001), whereas BP in females PT-4a12 mice was not significantly different from WT (118±2 vs. 117±2 mmHg; p=0.98). In male PT-4a12 mice, BP decreased when transitioned from a control salt (0.4%) to a low-salt diet (0.075%) from 135±4 to 120±6 mmHg (p<0.01) and increased to 153±5 mmHg (p<0.05) when placed on a high-salt diet (4%). Female PT-4a12 mice did not show changes in BP on either low- or high-salt diet. In conclusion, the expression of Cyp4a12 driven by the PEPCK promoter is sex-specific probably due to its X-linkage. The salt-sensitive hypertension seen in PT-4a12 male mice suggests a potential anti-natriuretic activity of 20-HETE that needs to be further explored.

7.
Am J Physiol Heart Circ Physiol ; 318(4): H985-H993, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32167781

ABSTRACT

The roles of ACE-independent ANG II production via chymase and therapeutic potential of epoxyeicosatrienoic acids (EETs) in fructose-induced metabolic syndrome (MetS) in the adolescent population remain elusive. Thus we tested the hypothesis that a high-fructose diet (HFD) in young rats elicits chymase-dependent increases in ANG II production and oxidative stress, responses that are reversible by 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU), an inhibitor of soluble epoxide hydrolase (sEH) that metabolizes EETs. Three groups of weanling rats (21-day-old) were fed a normal diet, 60% HFD, and HFD with TPPU, respectively, for 30 days. HFD rats developed MetS, characterized by hyperglycemia, hyperinsulinemia, and hypertension and associated with decreases in cardiac output and stroke volume and loss of nitric oxide (NO) modulation of myocardial oxygen consumption; all impairments were normalized by TPPU that significantly elevated circulating 11,12-EET, a major cardiac EET isoform. In the presence of comparable cardiac angiotensin-converting enzyme (ACE) expression/activity among the three groups, HFD rats exhibited significantly greater chymase-dependent ANG II formation in hearts, as indicated by an augmented cardiac chymase content as a function of enhanced mast cell degranulation. The enhanced chymase-dependent ANG II production was paralleled with increases in ANG II type 1 receptor (AT1R) expression and NADPH oxidase (Nox)-induced superoxide, alterations that were significantly reversed by TPPU. Conversely, HFD-induced downregulation of cardiac ACE2, followed by a lower Ang-(1-7) level displayed in an TPPU-irreversible manner. In conclusion, HFD-driven adverse chymase/ANG II/Nox/superoxide signaling in young rats was prevented by inhibition of sEH via, at least in part, an EET-mediated stabilization of mast cells, highlighting chymase and sEH as therapeutic targets during treatment of MetS.NEW & NOTEWORTHY As the highest fructose consumers, the adolescent population is highly susceptible to the metabolic syndrome, where increases in mast cell chymase-dependent formation of ANG II, ensued by cardiometabolic dysfunction, are reversible in response to inhibition of soluble epoxide hydrolase (sEH). This study highlights chymase and sEH as therapeutic targets and unravels novel avenues for the development of optimal strategies for young patients with fructose-induced metabolic syndrome.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Angiotensin II/metabolism , Chymases/metabolism , Fructose/adverse effects , Heart Diseases/metabolism , Metabolic Syndrome/complications , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cardiac Output , Cells, Cultured , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Heart/drug effects , Heart/physiopathology , Heart Diseases/drug therapy , Heart Diseases/etiology , Heart Rate , Male , Metabolic Syndrome/etiology , Myocardium/metabolism , Oxidative Stress , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Rats , Rats, Sprague-Dawley
9.
Pulm Circ ; 9(4): 2045894019895947, 2019.
Article in English | MEDLINE | ID: mdl-31908769

ABSTRACT

Epoxyeicosatrienoic acids (EETs) are synthesized from arachidonic acid by CYP/epoxygenase and metabolized by soluble epoxide hydrolase (sEH). Roles of EETs in hypoxia-induced pulmonary hypertension (HPH) remain elusive. The present study aimed to investigate the underlying mechanisms, by which EETs potentiate HPH. Experiments were conducted on sEH knockout (sEH-KO) and wild type (WT) mice after exposure to hypoxia (10% oxygen) for three weeks. In normal/normoxic conditions, WT and sEH-KO mice exhibited comparable pulmonary artery acceleration time (PAAT), ejection time (ET), PAAT/ET ratio, and velocity time integral (VTI), along with similar right ventricular systolic pressure (RVSP). Chronic hypoxia significantly reduced PAAT, ET, and VTI, coincided with an increase in RVSP; these impairments were more severe in sEH-KO than WT mice. Hypoxia elicited downregulation of sEH and upregulation of CYP2C9 accompanied with elevation of CYP-sourced superoxide, leading to enhanced pulmonary EETs in hypoxic mice with significantly higher levels in sEH-KO mice. Isometric tension of isolated pulmonary arteries was recorded. In addition to downregulation of eNOS-induced impairment of vasorelaxation to ACh, HPH mice displayed upregulation of thromboxane A2 (TXA2) receptor, paralleled with enhanced pulmonary vasocontraction to a TXA2 analog (U46619) in an sEH-KO predominant manner. Inhibition of COX-1 or COX-2 significantly prevented the enhancement by ∼50% in both groups of vessels, and the remaining incremental components were eliminated by scavenging of superoxide with Tiron. In conclusion, hypoxia-driven increases in EETs, intensified COXs/TXA2 signaling, great superoxide sourced from activated CYP2C9, and impaired NO bioavailability work in concert, to potentiate HPH development.

10.
Antioxid Redox Signal ; 31(10): 752-769, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30403147

ABSTRACT

Significance: This review considers how some systems controlling pulmonary vascular function are potentially regulated by redox processes to examine how and why conditions such as prolonged hypoxia, pathological mediators, and other factors promoting vascular remodeling contribute to the development of pulmonary hypertension (PH). Recent Advances and Critical Issues: Aspects of vascular remodeling induction mechanisms described are associated with shifts in glucose metabolism through the pentose phosphate pathway and increased cytosolic NADPH generation by glucose-6-phosphate dehydrogenase, increased glycolysis generation of cytosolic NADH and lactate, mitochondrial dysfunction associated with superoxide dismutase-2 depletion, changes in reactive oxygen species and iron metabolism, and redox signaling. Future Directions: The regulation and impact of hypoxia-inducible factor and the function of cGMP-dependent and redox regulation of protein kinase G are considered for their potential roles as key sensors and coordinators of redox and metabolic processes controlling the progression of vascular pathophysiology in PH, and how modulating aspects of metabolic and redox regulatory systems potentially function in beneficial therapeutic approaches.


Subject(s)
Blood Vessels/metabolism , Animals , Blood Vessels/physiology , Blood Vessels/physiopathology , Cyclic GMP/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glycolysis , Humans , Hypertension, Pulmonary/metabolism , Lactic Acid/metabolism , NAD/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
11.
PLoS One ; 13(7): e0200307, 2018.
Article in English | MEDLINE | ID: mdl-29985945

ABSTRACT

Ferrochelatase (FECH) is an enzyme necessary for heme synthesis, which is essential for maintaining normal functions of endothelial nitric oxide synthase (eNOS) and soluble guanylyl cyclase (sGC). We tested the hypothesis that inhibition of vascular FECH to attenuate heme synthesis downregulates eNOS and sGC expression, resulting in impaired NO/cGMP-dependent relaxation. To this end, isolated bovine coronary arteries (BCAs) were in vitro incubated without (as controls) or with N-methyl protoporphyrin (NMPP; 10(-5)-10(-7)M; a selective FECH antagonist) for 24 and 72 hours respectively. Tissue FECH activity, heme, nitrite/NO and superoxide levels were sequentially measured. Protein expression of FECH, eNOS and sGC was detected by western blot analysis. Vascular responses to various vasoactive agents were evaluated via isometric tension studies. Treatment of BCAs with NMPP initiated a time- and dose-dependent attenuation of FECH activity without changes in its protein expression, followed by significant reduction in the heme level. Moreover, ACh-induced relaxation and ACh-stimulated release of NO were significant reduced, associated with suppression of eNOS protein expression in NMPP-treated groups. Decreased relaxation to NO donor spermine-NONOate reached the statistical significance in BCAs incubated with NMPP for 72 hours, concomitantly with downregulation of sGCß1 expression that was independent of heat shock protein 90 (HSP90), nor did it significantly affect BCA relaxation caused by BAY 58-2667 that activates sGC in the heme-deficiency. Neither vascular responses to non-NO/sGC-mediators nor production of superoxide was affected by NMPP-treatment. In conclusion, deletion of vascular heme production via inhibiting FECH elicits downregulation of eNOS and sGC expression, leading to an impaired NO-mediated relaxation in an oxidative stress-independent manner.


Subject(s)
Coronary Vessels/drug effects , Cyclic GMP/metabolism , Ferrochelatase/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects , Soluble Guanylyl Cyclase/metabolism , Acetylcholine/pharmacology , Animals , Cattle , Coronary Vessels/metabolism , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/metabolism , Protoporphyrins/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
12.
J Am Heart Assoc ; 7(8)2018 04 13.
Article in English | MEDLINE | ID: mdl-29654195

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage is a serious clinical condition that impairs local cerebral blood flow perfusion and consequently initiates neuronal dysfunction. Pressure-sensitive myogenic vasomotor regulation is an important mechanism involved in the regulation of cerebral blood flow. We hypothesized that extravascular hemolyzed blood enhances arteriolar myogenic constriction, which in vivo may contribute to the reduction of local cerebral blood flow after subarachnoid hemorrhage. METHODS AND RESULTS: Arterioles isolated from the middle cerebral artery (MCA arterioles) of mice were cannulated in a perfusion chamber. Arteriolar diameters in response to step increases in intraluminal pressure (20-120 mm Hg) were measured in various experimental conditions. In response to increases in intraluminal pressure, all MCA arterioles exhibited myogenic vasoconstrictions. Compared with controls, the pressure-induced constriction was significantly enhanced in arterioles (in vitro) exposed to extravascular hemolyzed blood or different concentrations of extracellular erythrocyte lysate (1%, 10%, and 20%) for different exposure durations (1-6 hours). The magnitude of enhancement was proportional to the lysate concentration and exposure duration. In in vivo experiments, 10 µL of autologous blood lysate were injected into the mouse subarachnoid space on the surface of the left MCA. Two hours later, MCA arterioles were isolated and left MCA arterioles displayed enhanced myogenic responses compared with the right MCA. The enhanced myogenic response was prevented by scavenge of superoxide in both in vitro and in vivo experiments. CONCLUSIONS: Extravascular hemolyzed blood, perhaps by promoting vascular production of superoxide, augments myogenic constriction of cerebral arterioles, which plays a crucial role in the subarachnoid hemorrhage-induced cerebral ischemia.


Subject(s)
Arterioles/physiopathology , Blood Pressure/physiology , Cerebrovascular Circulation/physiology , Muscle, Smooth, Vascular/physiopathology , Subarachnoid Hemorrhage/physiopathology , Vasoconstriction/physiology , Animals , Disease Models, Animal , Hemolysis , Male , Mice , Mice, Inbred C57BL , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/diagnosis
13.
Proc Natl Acad Sci U S A ; 115(3): 613-618, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29295935

ABSTRACT

To elucidate molecular mechanisms responsible for the sexually dimorphic phenotype of soluble epoxide hydrolase (sEH) expression, we tested the hypothesis that female-specific down-regulation of sEH expression is driven by estrogen-dependent methylation of the Ephx2 gene. Mesenteric arteries isolated from male, female, ovariectomized female (OV), and OV with estrogen replacement (OVE) mice, as well as the human cell line (HEK293T) were used. Methylation-specific PCR and bisulfite genomic sequencing analysis indicate significant increases in DNA/CG methylation in vessels of female and OVE compared with those of male and OV mice. The same increase in CG methylation was also observed in male vessels incubated with a physiological concentration of 17ß-estradiol (17ß-E2) for 48 hours. All vessels that displayed increases in CG methylation were concomitantly associated with decreases in their Ephx2 mRNA and protein, suggesting a methylation-induced gene silencing. Transient transfection assays indicate that the activity of Ephx2 promoter-coding luciferase was significantly attenuated in HEK293T cells treated with 17ß-E2, which was prevented by additional treatment with an estrogen receptor antagonist (ICI). ChIP analysis indicates significantly reduced binding activities of transcription factors (including SP1, AP-1, and NF-κB with their binding elements located in the Ephx2 promoter) in vessels of female mice and human cells treated with 17ß-E2, responses that were prevented by ICI and Decitabine (DNA methyltransferase inhibitor), respectively. In conclusion, estrogen/estrogen receptor-dependent methylation of the promoter of Ephx2 gene silences sEH expression, which is involved in specific transcription factor-directed regulatory pathways.


Subject(s)
Epigenesis, Genetic , Epoxide Hydrolases/genetics , Estradiol/metabolism , Estrogens/metabolism , Animals , DNA Methylation , Epoxide Hydrolases/metabolism , Female , Gene Silencing , HEK293 Cells , Humans , Male , Mesenteric Arteries/metabolism , Mice , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Article in English | MEDLINE | ID: mdl-28827137

ABSTRACT

To test the hypothesis that VitC downregulates soluble epoxide hydrolase (sEH, responsible for converting EETs to DHETs) to stabilize tissue EETs, the heart, lung, liver, kidney, and mesenteric arteries isolated from normal rats were incubated with VitC (1000µM) for 72h, and tissue sEH expression, along with EET and DHET profiles were assessed. VitC caused significant reductions in sEH mRNA and protein content in the liver, heart and vessels, but had no effect on renal and pulmonary sEH expression, revealing a tissue-specific regulatory mechanism. The functional consequence of reduced sEH expression was validated by LC/MS/MS-based analysis, indicating that in VitC-treated tissues that displayed downregulation of sEH mRNA and protein expression, total DHETs were significantly lower, accompanied with a greater ratio of EETs/DHETs than those in VitC-untreated groups. Thus, VitC elicits a transcriptional downregulation of sEH in normal liver, heart, and vessels to reduce EET degradation and increase EET bioavailability.


Subject(s)
Ascorbic Acid/pharmacology , Epoxide Hydrolases/chemistry , Epoxide Hydrolases/metabolism , Animals , Epoxide Hydrolases/genetics , Gene Expression Regulation, Enzymologic/drug effects , Male , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Solubility
15.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L350-L359, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28450284

ABSTRACT

To test the hypothesis that epoxyeicosatrienoic acids (EETs) facilitate pulmonary responses to hypoxia, male wild-type (WT) and soluble-epoxide hydrolase knockout (sEH-KO) mice, and WT mice chronically fed a sEH inhibitor (t-TUCB; 1 mg·kg-1·day-1) were used. Right ventricular systolic pressure (RVSP) was recorded under control and hypoxic conditions. The control RVSP was comparable among all groups. However, hypoxia elicited increases in RVSP in all groups with predominance in sEH-KO and t-TUCB-treated mice. 14,15-EEZE (an EET antagonist) attenuated the hypoxia-induced greater elevation of RVSP in sEH-deficient mice, suggesting an EET-mediated increment. Exogenous 5,6-; 8,9-, or 14,15-EET (0.05 ng/g body wt) did not change RVSP in any conditions, but 11,12-EET enhanced RVSP under hypoxia. Isometric tension was recorded from pulmonary arteries isolated from WT and sEH-KO mice, vessels that behaved identically in their responsiveness to vasoactive agents and vessel stretch. Hypoxic pulmonary vasoconstriction (HPV, expressed as increases in hypoxic force) was significantly greater in vessels of sEH-KO than WT vessels; the enhanced component was inhibited by EEZE. Treatment of WT vessels with 11,12-EET enhanced HPV to the same level as sEH-KO vessels, confirming EETs as primary players. Inhibition of cyclooxygenases (COXs) significantly enhanced HPV in WT vessels, but attenuated HPV in sEH-KO vessels. Blocking/inhibiting COX-1, prostaglandin H2 (PGH2)/thromboxane A2 (TXA2) receptors and TXA synthase prevented the enhanced HPV in sEH-KO vessels but had no effects on WT vessels. In conclusion, an EET-dependent alteration in PG metabolism that favors the action of vasoconstrictor PGH2 and TXA2 potentiates HPV and hypoxia-induced elevation of RVSP in sEH-deficient mice.


Subject(s)
8,11,14-Eicosatrienoic Acid/pharmacology , Hypoxia/chemically induced , Prostaglandins/metabolism , Pulmonary Artery/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Animals , Blood Pressure/drug effects , Epoxide Hydrolases/pharmacology , Hypoxia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Artery/metabolism
16.
Am J Physiol Heart Circ Physiol ; 312(2): H223-H231, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27815252

ABSTRACT

Age-dependent alteration of the renin-angiotensin system (RAS) and generation of angiotensin II (Ang II) are well documented. By contrast, RAS-independent generation of Ang II in aging and its responses to exercise have not been explored. To this end, we examined the effects of chymase, a secretory serine protease, on the angiotensin-converting enzyme (ACE)-independent conversion of Ang I to Ang II. We hypothesized that age-dependent alteration of cardiac Ang II formation is chymase dependent in nature and is prevented by exercise training. Experiments were conducted on hearts isolated from young (3 mo), aged sedentary (24 mo), and aged rats chronically exercised on a treadmill. In the presence of low Ang I levels and downregulation of ACE expression/activity, cardiac Ang II levels were significantly higher in aged than young rats, suggesting an ACE-independent response. Aged hearts also displayed significantly increased chymase expression and activity, as well as upregulation of tryptase, a biological marker of mast cells, confirming a mast cell-sourced increase in chymase. Coincidently, cardiac superoxide produced from NADPH oxidase (Nox) was significantly enhanced in aged rats and was normalized by exercise. Conversely, a significant reduction in cardiac expression of ACE2 followed by lower Ang 1-7 levels and downregulation of the Mas receptor (binding protein of Ang 1-7) in aged rats were completely reversed by exercise. In conclusion, local formation of Ang II is increased in aged hearts, and chymase is primarily responsible for this increase. Chronic exercise is able to normalize the age-dependent alterations via compromising chymase/Ang II/angiotensin type 1 receptor/Nox actions while promoting ACE2/Ang 1-7/MasR signaling. NEW & NOTEWORTHY: Aging increases angiotensin-converting enzyme (ACE)-independent production of cardiac angiotensin II (Ang II), a response that is driven by chymase in an exercise-reversible manner. These findings highlight chymase, in addition to ACE, as an important therapeutic target in the treatment and prevention of Ang II-induced deterioration of cardiac function in the elderly.


Subject(s)
Aging/metabolism , Angiotensin II/metabolism , Angiotensin I/metabolism , Chymases/metabolism , Myocardium/metabolism , Peptide Fragments/metabolism , Physical Conditioning, Animal , Angiotensin-Converting Enzyme 2 , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , NADPH Oxidases/metabolism , Peptidyl-Dipeptidase A/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Rats , Rats, Inbred F344 , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Superoxides/metabolism
17.
Physiol Rep ; 4(12)2016 Jun.
Article in English | MEDLINE | ID: mdl-27354541

ABSTRACT

Epoxyeicosatrienoic acids (EETs) are cardioprotective mediators metabolized by soluble epoxide hydrolase (sEH) to form corresponding diols (DHETs). As a sex-susceptible target, sEH is involved in the sexually dimorphic regulation of cardiovascular function. Thus, we hypothesized that the female sex favors EET-mediated potentiation of cardiac function via downregulation of sEH expression, followed by upregulation of peroxisome proliferator-activated receptors (PPARs). Hearts were isolated from male (M) and female (F) wild-type (WT) and sEH-KO mice, and perfused with constant flow at different preloads. Basal coronary flow required to maintain the perfusion pressure at 100 mmHg was significantly greater in females than males, and sEH-KO than WT mice. All hearts displayed a dose-dependent decrease in coronary resistance and increase in cardiac contractility, represented as developed tension in response to increases in preload. These responses were also significantly greater in females than males, and sEH-KO than WT 14,15-EEZE abolished the sex-induced (F vs. M) and transgenic model-dependent (KO vs. WT) differences in the cardiac contractility, confirming an EET-driven response. Compared with M-WT controls, F-WT hearts expressed downregulation of sEH, associated with increased EETs and reduced DHETs, a pattern comparable to that observed in sEH-KO hearts. Coincidentally, F-WT and sEH-KO hearts exhibited increased PPARα expression, but comparable expression of eNOS, PPARß, and EET synthases. In conclusion, female-specific downregulation of sEH initiates an EET-dependent adaptation of cardiac function, characterized by increased coronary flow via reduction in vascular resistance, and promotion of cardiac contractility, a response that could be further intensified by PPARα.


Subject(s)
8,11,14-Eicosatrienoic Acid/metabolism , Adaptation, Physiological , Epoxide Hydrolases/metabolism , Heart/physiology , Myocardial Contraction , Peroxisome Proliferator-Activated Receptors/metabolism , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Animals , Coronary Circulation , Epoxide Hydrolases/genetics , Female , Male , Mice , Myocardium/metabolism , Sex Factors
18.
Am J Physiol Heart Circ Physiol ; 310(11): H1448-54, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27016584

ABSTRACT

Epoxyeicosatrienoic acids (EETs) are metabolites of arachidonic acid via CYP/epoxygenases, which are catabolized by soluble epoxide hydrolase (sEH) and known to possess cardioprotective properties. To date, the role of sEH in the modulation of pressure-induced myogenic response/constriction in coronary arteries, an important regulatory mechanism in the coronary circulation, and the issue as to whether the disruption of the sEH gene affects the myogenic response sex differentially have never been addressed. To this end, experiments were conducted on male (M) and female (F) wild-type (WT) and sEH-knockout (KO) mice. Pressure-diameter relationships were assessed in isolated and cannulated coronary arteries. All vessels constricted in response to increases in intraluminal pressure from 60 to 120 mmHg. Myogenic vasoconstriction was significantly attenuated, expressed as an upward shift in the pressure-diameter curve of vessels, associated with higher cardiac EETs in M-KO, F-WT, and F-KO mice compared with M-WT controls. Blockade of EETs via exposure of vessels to 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE) prevented the attenuated myogenic constriction in sEH-KO mice. In the presence of 14,15-EEZE, pressure-diameter curves of females presented an upward shift from those of males, exhibiting a sex-different phenotype. Additional administration of N(ω)-nitro-l-arginine methyl ester eliminated the sex difference in myogenic responses, leading to four overlapped pressure-diameter curves. Cardiac sEH was downregulated in F-WT compared with M-WT mice, whereas expression of endothelial nitric oxide synthase and CYP4A (20-HETE synthase) was comparable among all groups. In summary, in combination with NO, the increased EET bioavailability as a function of genetic deletion and/or downregulation of sEH accounts for the female-favorable attenuation of pressure-induced vasoconstriction.


Subject(s)
Arachidonic Acids/metabolism , Coronary Vessels/metabolism , Nitric Oxide/metabolism , Vasoconstriction , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Arachidonic Acids/antagonists & inhibitors , Arterial Pressure , Coronary Vessels/drug effects , Cytochrome P-450 CYP4A/metabolism , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/deficiency , Epoxide Hydrolases/genetics , Female , Genotype , Hydroxyeicosatetraenoic Acids/metabolism , In Vitro Techniques , Male , Mechanotransduction, Cellular , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phenotype , Sex Factors , Vasoconstriction/drug effects
19.
Am J Hypertens ; 29(5): 598-604, 2016 May.
Article in English | MEDLINE | ID: mdl-26304959

ABSTRACT

OBJECTIVE: The biological role of epoxyeicosatrienoic acids (EETs) in the regulation of pulmonary circulation is currently under debate. We hypothesized that EETs initiate increases in right ventricular systolic pressure (RVSP) via perhaps, pulmonary vasoconstriction. METHODS: Mice were anesthetized with isoflurane. Three catheters, inserted into the left jugular vein, the left carotid artery, and the right jugular vein, were used for infusing EETs, monitoring blood pressure (BP), and RVSP respectively. BP and RVSP were continuously recorded at basal conditions, in response to administration of 4 regioisomeric EETs (5,6-EET; 8,9-EET; 11,12-EET, and 14,15-EET; 1, 2, 5 and 10 ng/g body weight (BW) for each EET), and during exposure of mice to hypoxia. RESULTS: All 4 EETs initiated dose-dependent increases in RVSP, though reduced BP. 11,12-EET elicited the greatest increment in RVSP among all EET isoforms. To clarify the direct elevation of RVSP in a systemic BP-independent manner, equivalent amounts of 14,15-EET were injected over 1 and 2 minutes respectively. One-minute injection of 14,15-EET elicited significantly faster and greater increases in RVSP than the 2-minute injection, whereas their BP changes were comparable. Additionally, direct injection of low doses of 14,15-EET (0.1, 0.2, 0.5, and 1 ng/g BW) into the right ventricle caused significant increases in RVSP without effects on BP, confirming that systemic vasodilation-induced increases in venous return are not the main cause for the increased RVSP. Acute exposure of mice to hypoxia significantly elevated RVSP, as well as 14,15-EET-induced increases in RVSP. CONCLUSIONS: EETs directly elevate RVSP, a response that may play an important role in the development of hypoxia-induced pulmonary hypertension (PH).


Subject(s)
8,11,14-Eicosatrienoic Acid/toxicity , Arterial Pressure/drug effects , Hypertension, Pulmonary/chemically induced , Pulmonary Artery/drug effects , 8,11,14-Eicosatrienoic Acid/administration & dosage , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Infusions, Intravenous , Male , Mice, Inbred C57BL , Pulmonary Artery/physiopathology , Time Factors , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects
20.
Am J Physiol Lung Cell Mol Physiol ; 309(12): L1478-86, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26498250

ABSTRACT

We tested the hypothesis that suppression of epoxyeicosatrienoic acid (EET) metabolism via genetic knockout of the gene for soluble epoxide hydrolase (sEH-KO), or female-specific downregulation of sEH expression, plays a role in the potentiation of pulmonary hypertension. We used male (M) and female (F) wild-type (WT) and sEH-KO mice; the latter have high pulmonary EETs. Right ventricular systolic pressure (RVSP) and mean arterial blood pressure (MABP) in control and in response to in vivo administration of U46619 (thromboxane analog), 14,15-EET, and 14,15-EEZE [14,15-epoxyeicosa-5(z)-enoic acid; antagonist of EETs] were recorded. Basal RVSP was comparable among all groups of mice, whereas MABP was significantly lower in F-WT than M-WT mice and further reduced predominantly in F-KO compared with M-KO mice. U46619 dose dependently increased RVSP and MABP in all groups of mice. The increase in RVSP was significantly greater and coincided with smaller increases in MABP in M-KO and F-WT mice compared with M-WT mice. In F-KO mice, the elevation of RVSP by U46619 was even higher than in M-KO and F-WT mice, associated with the least increase in MABP. 14,15-EEZE prevented the augmentation of U46619-induced elevation of RVSP in sEH-KO mice, whereas 14,15-EET-induced pulmonary vasoconstriction was comparable in all groups of mice. sEH expression in the lungs was reduced, paralleled with higher levels of EETs in F-WT compared with M-WT mice. In summary, EETs initiate pulmonary vasoconstriction but act as vasodilators systemically. High pulmonary EETs, as a function of downregulation or deletion of sEH, potentiate U46619-induced increases in RVSP in a female-susceptible manner.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arterial Pressure/drug effects , Arterial Pressure/physiology , Epoxide Hydrolases/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Female , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Male , Mice , Mice, Knockout , Sex Characteristics , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...