Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Hypertension ; 81(5): 1008-1020, 2024 May.
Article in English | MEDLINE | ID: mdl-38426347

ABSTRACT

After half a century of evidence suggesting the existence of mineralocorticoid receptors (MR) in the vasculature, the advent of technology to specifically knockout the MR from smooth muscle cells (SMCs) in mice has elucidated contributions of SMC-MR to cardiovascular function and disease, independent of the kidney. This review summarizes the latest understanding of the molecular mechanisms by which SMC-MR contributes to (1) regulation of vasomotor function and blood pressure to contribute to systemic and pulmonary hypertension; (2) vascular remodeling in response to hypertension, vascular injury, obesity, and aging, and the impact on vascular calcification; and (3) cardiovascular pathologies including aortic aneurysm, heart valve dysfunction, and heart failure. Data are reviewed from in vitro studies using SMCs and in vivo findings from SMC-specific MR-knockout mice that implicate target genes and signaling pathways downstream of SMC-MR. By regulating expression of the L-type calcium channel subunit Cav1.2 and angiotensin II type-1 receptor, SMC-MR contributes to myogenic tone and vasoconstriction, thereby contributing to systemic blood pressure. MR activation also promotes SMC proliferation, migration, production and degradation of extracellular matrix, and osteogenic differentiation by regulating target genes including connective tissue growth factor, osteopontin, bone morphogenetic protein 2, galectin-3, and matrix metallopeptidase-2. By these mechanisms, SMC-MR promotes disease progression in models of aging-associated vascular stiffness, vascular calcification, mitral and aortic valve disease, pulmonary hypertension, and heart failure. While rarely tested, when sexes were compared, the mechanisms of SMC-MR-mediated disease were sexually dimorphic. These advances support targeting SMC-MR-mediated mechanisms to prevent and treat diverse cardiovascular disorders.


Subject(s)
Heart Failure , Hypertension, Pulmonary , Vascular Calcification , Animals , Mice , Blood Pressure/physiology , Receptors, Mineralocorticoid/metabolism , Muscle, Smooth, Vascular/metabolism , Hypertension, Pulmonary/metabolism , Osteogenesis , Heart Failure/metabolism , Vascular Calcification/metabolism , Myocytes, Smooth Muscle/metabolism
2.
Arterioscler Thromb Vasc Biol ; 44(4): 946-953, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38450510

ABSTRACT

BACKGROUND: Women with a history of preeclampsia have evidence of premature atherosclerosis and increased risk of myocardial infarction and stroke compared with women who had a normotensive pregnancy. Whether this is due to common risk factors or a direct impact of prior preeclampsia exposure has never been tested in a mouse atherosclerosis model. METHODS: Pregnant LDLR-KO (low-density lipoprotein receptor knockout; n=35) female mice were randomized in midgestation to sFlt1 (soluble fms-like tyrosine kinase 1)-expressing adenovirus or identical control adenovirus. Postpartum, mice were fed high-fat diet for 8 weeks to induce atherogenesis. Comparison between the control and preeclampsia models was made for metabolic parameters, atherosclerosis burden and composition by histology, plaque inflammation by flow cytometry, and aortic cytokines and inflammatory markers using a cytokine array. RESULTS: In pregnant LDLR-KO mice, sFlt1 adenovirus significantly induced serum sFlt1, blood pressure, renal endotheliosis, and decreased pup viability. After 8 weeks of postpartum high fat feeding, body weight, fasting glucose, plasma cholesterol, HDL (high-density lipoprotein), and LDL (low-density lipoprotein) were not significantly different between groups with no change in aortic root plaque size, lipid content, or necrotic core area. Flow cytometry demonstrated significantly increased CD45+ aortic arch leukocytes and CD3+T cells and aortic lysate contained more CCL (CC motif chemokine ligand) 22 and fetuin A and decreased expression of IGFBP6 (insulin-like growth factor-binding protein 6) and CCL21 in preeclampsia-exposed mice compared with controls. CONCLUSIONS: In atherogenic LDLR-KO mice, exposure to sFlt1-induced preeclampsia during pregnancy increases future atherosclerotic plaque inflammation, supporting the concept that preeclampsia directly exacerbates atherosclerotic inflammation independent of preexisting risk factors. This mechanism may contribute to ischemic vascular disease in women after preeclampsia pregnancy.


Subject(s)
Aortic Diseases , Atherosclerosis , Plaque, Atherosclerotic , Pre-Eclampsia , Humans , Female , Animals , Mice , Vascular Endothelial Growth Factor Receptor-1/genetics , Aortic Diseases/genetics , Mice, Knockout , Atherosclerosis/genetics , Inflammation/metabolism , Lipoproteins, LDL/metabolism , Receptors, LDL/genetics , Cytokines , Mice, Inbred C57BL
3.
Antioxidants (Basel) ; 12(12)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38136156

ABSTRACT

Background: Preeclampsia (PE) is a hypertensive disorder of pregnancy that is associated with substantial morbidity and mortality for the mother and fetus. Reduced nitric oxide bioavailability and oxidative stress contribute to the maternal and fetal pathophysiology of PE. In this study, we evaluated the efficacy of a novel dual-function nitric oxide donor/redox modulator, AKT-1005, in reducing PE symptoms in a mouse model of PE. Method: The potential therapeutic effect of AKT-1005 was tested in an animal model of Ad.sFlt-1-induced hypertension, proteinuria and glomerular endotheliosis, a model of PE. Pregnant Ad.sFlt-1-overexpressing CD1 mice were randomized into groups administered AKT-1005 (20 mg/kg) or a vehicle using a minipump on gd11 of pregnancy, and the impact on blood pressure and renal and placental damage were assessed. Results: In healthy female mice, ex vivo treatment of resistance vessels with AKT-1005 induced vasorelaxation, and 6 days of treatment in vivo did not significantly alter blood pressure with or without pregnancy. When given for 6 days during pregnancy along with Ad.sFlt-1-induced PE, AKT-1005 significantly increased plasma nitrate levels and reduced hypertension, renal endotheliosis and plasma cystatin C. In the placenta, AKT-1005 improved placental function, with reduced oxidative stress and increased endothelial angiogenesis, as measured by CD31 staining. As such, AKT-1005 treatment attenuated the Ad.sFlt-1-induced increase in placental and free plasma soluble endoglin expression. Conclusions: These data suggest that AKT-1005 significantly attenuates the sFlt-1-induced PE phenotypes by inhibiting oxidative stress, the anti-angiogenic response, and increasing NO bioavailability. Additional research is warranted to investigate the role of AKT-1005 as a novel therapeutic agent for vascular disorders such as preeclampsia.

4.
Circ Res ; 132(6): 674-689, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36815487

ABSTRACT

BACKGROUND: Preeclampsia is a syndrome of high blood pressure (BP) with end organ damage in late pregnancy that is associated with high circulating soluble VEGF receptor (sFlt1 [soluble Fms-like tyrosine kinase 1]). Women exposed to preeclampsia have a substantially increased risk of hypertension after pregnancy, but the mechanism remains unknown, leaving a missed interventional opportunity. After preeclampsia, women have enhanced sensitivity to hypertensive stress. Since smooth muscle cell mineralocorticoid receptors (SMC-MR) are activated by hypertensive stimuli, we hypothesized that high sFlt1 exposure in pregnancy induces a postpartum state of enhanced SMC-MR responsiveness. METHODS: Postpartum BP response to high salt intake was studied in women with prior preeclampsia. MR transcriptional activity was assessed in vitro in sFlt1-treated SMC by reporter assays and PCR. Preeclampsia was modeled by transient sFlt1 expression in pregnant mice. Two months post-partum, mice were exposed to high salt and then to AngII (angiotensin II) and BP and vasoconstriction were measured. RESULTS: Women exposed to preeclampsia had significantly enhanced salt sensitivity of BP verses those with a normotensive pregnancy. sFlt1 overexpression during pregnancy in mice induced elevated BP and glomerular endotheliosis, which resolved post-partum. The sFlt1 exposed post-partum mice had significantly increased BP response to 4% salt diet and to AngII infusion. In vitro, SMC-MR transcriptional activity in response to aldosterone or AngII was significantly increased after transient exposure to sFlt1 as was aldosterone-induced expression of AngII type 1 receptor. Post-partum, SMC-MR-KO mice were protected from the enhanced response to hypertensive stimuli after preeclampsia. Mechanistically, preeclampsia mice exposed to postpartum hypertensive stimuli develop enhanced aortic stiffness, microvascular myogenic tone, AngII constriction, and AngII type 1 receptor expression, all of which were prevented in SMC-MR-KO littermates. CONCLUSIONS: These data support that sFlt1-induced vascular injury during preeclampsia produces a persistent state of enhanced sensitivity of SMC-MR to activation. This contributes to postpartum hypertension in response to common stresses and supports testing of MR antagonism to mitigate the increased cardiovascular risk in women after PE.


Subject(s)
Hypertension , Pre-Eclampsia , Humans , Pregnancy , Female , Mice , Animals , Pre-Eclampsia/etiology , Pre-Eclampsia/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Receptors, Mineralocorticoid/genetics , Aldosterone , Muscle, Smooth/metabolism
5.
Am J Physiol Heart Circ Physiol ; 323(6): H1212-H1220, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36306211

ABSTRACT

The fat mass and obesity gene (FTO) is a N6-methyladenosine RNA demethylase that was initially linked by Genome-wide association studies to increased rates of obesity. Subsequent studies have revealed multiple mass-independent effects of the gene, including cardiac myocyte contractility. We created a mouse with a conditional and inducible smooth muscle cell deletion of Fto (Myh11 Cre+ Ftofl/fl) and did not observe any changes in mouse body mass or mitochondrial metabolism. However, the mice had significantly decreased blood pressure (hypotensive), despite increased heart rate and sodium, and significantly increased plasma renin. Remarkably, the third-order mesenteric arteries from these mice had almost no myogenic tone or capacity to constrict to smooth muscle depolarization or phenylephrine. Microarray analysis from Fto-/--isolated smooth muscle cells demonstrated a significant decrease in serum response factor (Srf) and the downstream effectors Acta2, Myocd, and Tagln; this was confirmed in cultured human coronary arteries with FTO siRNA. We conclude Fto is an important component to the contractility of smooth muscle cells.NEW & NOTEWORTHY We show a key role for the fat mass obesity (FTO) gene in regulating smooth muscle contractility, possibly by methylation of serum response factor (Srf).


Subject(s)
Genome-Wide Association Study , Serum Response Factor , Mice , Humans , Animals , Serum Response Factor/genetics , Myocytes, Smooth Muscle/metabolism , Obesity/genetics , Muscle Contraction , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism
7.
Sci Signal ; 14(672)2021 03 02.
Article in English | MEDLINE | ID: mdl-33653920

ABSTRACT

The endothelial cell barrier regulates the passage of fluid between the bloodstream and underlying tissues, and barrier function impairment exacerbates the severity of inflammatory insults. To understand how inflammation alters vessel permeability, we studied the effects of the proinflammatory cytokine TNFα on transendothelial permeability and electrophysiology in ex vivo murine veins and arteries. We found that TNFα specifically decreased the barrier function of venous endothelium without affecting that of arterial endothelium. On the basis of RNA expression profiling and protein analysis, we found that claudin-11 (CLDN11) was the predominant claudin in venous endothelial cells and that there was little, if any, CLDN11 in arterial endothelial cells. Consistent with a difference in claudin composition, TNFα increased the permselectivity of Cl- over Na+ in venous but not arterial endothelium. The vein-specific effects of TNFα also required the activation of Pannexin 1 (Panx1) channels and the CD39-mediated hydrolysis of ATP to adenosine, which subsequently stimulated A2A adenosine receptors. Moreover, the increase in vein permeability required the activation of the Ca2+ channel TRPV4 downstream of Panx1 activation. Panx1-deficient mice resisted the pathologic effects of sepsis induced by cecal ligation and puncture on life span and lung vascular permeability. These data provide a targetable pathway with the potential to promote vein barrier function and prevent the deleterious effects of vascular leak in response to inflammation.


Subject(s)
Connexins , Endothelial Cells , Nerve Tissue Proteins , Tumor Necrosis Factor-alpha , Animals , Capillary Permeability , Connexins/genetics , Connexins/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Permeability , TRPV Cation Channels/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Circ Heart Fail ; 14(2): e007279, 2021 02.
Article in English | MEDLINE | ID: mdl-33517669

ABSTRACT

BACKGROUND: Mineralocorticoid receptor (MR) antagonists decrease heart failure (HF) hospitalization and mortality, but the mechanisms are unknown. Preclinical studies reveal that the benefits on cardiac remodeling and dysfunction are not completely explained by inhibition of MR in cardiomyocytes, fibroblasts, or endothelial cells. The role of MR in smooth muscle cells (SMCs) in HF has never been explored. METHODS: Male mice with inducible deletion of MR from SMCs (SMC-MR-knockout) and their MR-intact littermates were exposed to HF induced by 27-gauge transverse aortic constriction versus sham surgery. HF phenotypes and mechanisms were measured 4 weeks later using cardiac ultrasound, intracardiac pressure measurements, exercise testing, histology, cardiac gene expression, and leukocyte flow cytometry. RESULTS: Deletion of MR from SMC attenuated transverse aortic constriction-induced HF with statistically significant improvements in ejection fraction, cardiac stiffness, chamber dimensions, intracardiac pressure, pulmonary edema, and exercise capacity. Mechanistically, SMC-MR-knockout protected from adverse cardiac remodeling as evidenced by decreased cardiomyocyte hypertrophy and fetal gene expression, interstitial and perivascular fibrosis, and inflammatory and fibrotic gene expression. Exposure to pressure overload resulted in a statistically significant decline in cardiac capillary density and coronary flow reserve in MR-intact mice. These vascular parameters were improved in SMC-MR-knockout mice compared with MR-intact littermates exposed to transverse aortic constriction. CONCLUSIONS: These results provide a novel paradigm by which MR inhibition may be beneficial in HF by blocking MR in SMC, thereby improving cardiac blood supply in the setting of pressure overload-induced hypertrophy, which in turn mitigates the adverse cardiac remodeling that contributes to HF progression and symptoms.


Subject(s)
Heart Failure/genetics , Myocytes, Smooth Muscle/metabolism , Receptors, Mineralocorticoid/genetics , Ventricular Remodeling/genetics , Animals , Aorta/surgery , Arterial Pressure , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Constriction, Pathologic , Disease Models, Animal , Echocardiography , Gene Knockout Techniques , Heart Failure/diagnostic imaging , Heart Failure/pathology , Heart Failure/physiopathology , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/physiology
9.
Am J Physiol Heart Circ Physiol ; 318(5): H1041-H1048, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32196361

ABSTRACT

Discrete calcium signals within the vascular endothelium decrease with age and contribute to impaired endothelial-dependent vasodilation. Calreticulin (Calr), a multifunctional calcium binding protein and endoplasmic reticulum (ER) chaperone, can mediate calcium signals and vascular function within the endothelial cells (ECs) of small resistance arteries. We found Calr protein expression significantly decreases with age in mesenteric arteries and examined the functional role of EC Calr in vasodilation and calcium mobilization in the context of aging. Third-order mesenteric arteries from mice with or without EC Calr knockdown were examined for calcium signals and constriction to phenylephrine (PE) or vasodilation to carbachol (CCh) after 75 wk of age. PE constriction in aged mice with or without EC Calr was unchanged. However, calcium signals and vasodilation to endothelial-dependent agonist carbachol were significantly impaired in aged EC Calr knockdown mice. Ex vivo incubation of arteries with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) significantly improved vasodilation in mice lacking EC Calr. Our data suggests diminished vascular Calr expression with age can contribute to the detrimental effects of aging on endothelial calcium regulation and vasodilation.NEW & NOTEWORTHY Calreticulin (Calr) is responsible for key physiological processes in endoplasmic reticulum, especially in aging tissue. In particular, endothelial Calr is crucial to vascular function. In this study, we deleted Calr from the endothelium and aged the mice up to 75 wk to examine changes in vascular function. We found two key differences: 1) calcium events in endothelium were severely diminished after muscarinic stimulation, which 2) corresponded with a dramatic decrease in muscarinic vasodilation. Remarkably, we were able to rescue the effect of Calr deletion on endothelial-dependent vasodilatory function using tauroursodeoxycholic acid (TUDCA), an inhibitor of endoplasmic reticulum stress that is currently in clinical trials.


Subject(s)
Aging/metabolism , Calreticulin/metabolism , Endothelium, Vascular/metabolism , Aging/physiology , Animals , Calcium Signaling , Calreticulin/genetics , Carbachol/pharmacology , Endothelium, Vascular/physiology , Gene Deletion , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Phenylephrine/pharmacology , Taurochenodeoxycholic Acid/pharmacology , Vasoconstrictor Agents/pharmacology , Vasodilation
10.
Front Physiol ; 11: 29, 2020.
Article in English | MEDLINE | ID: mdl-32116749

ABSTRACT

RATIONALE: Myoendothelial junctions (MEJs) within the fenestrae of the internal elastic lamina (IEL) are critical sites that allow for endothelial cell (EC) - vascular smooth muscle cell (VSMC) contact and communication. Vascular Notch signaling is a critical determinant of normal vasculogenesis and remodeling, and it regulates cell phenotype via contact between ECs and VSMCs. To date, no studies have linked Notch signaling to the MEJ despite it requiring cell-cell contact. Furthermore, very little is known about Notch in the adult coronary circulation or the localization of Notch signaling and activity within the mature intact blood vessel. OBJECTIVE: We tested the hypothesis that vascular Notch signaling between ECs and VSMCs occurs at MEJs. METHODS AND RESULTS: Notch receptor and ligand immunofluorescence was performed in human coronary EC and VSMC co-cultures across transwell inserts (in vitro MEJs) and in the intact mouse coronary circulation. Human coronary VSMC Notch activity induced by human coronary ECs at the in vitro MEJ was assessed using a CBF-luciferase construct. We observed Jagged1, Notch1, Notch2, and Notch3 expression within the in vitro and in vivo MEJs. We also demonstrated a 3-fold induction (p < 0.001) of human coronary VSMC Notch signaling by ECs at the in vitro MEJ, which was completely blocked by the Notch inhibitor, DAPT (p < 0.01). CONCLUSION: We demonstrate for the first time in mature blood vessels that Notch receptors and ligands are expressed within and are active at coronary MEJs, demonstrating a previously unrecognized mode of Notch signaling regulation between the endothelium and smooth muscle.

11.
Circ Res ; 126(2): 232-242, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31801409

ABSTRACT

RATIONALE: Increasing prevalence of obesity and its associated risk with cardiovascular diseases demands a better understanding of the contribution of different cell types within this complex disease for developing new treatment options. Previous studies could prove a fundamental role of FTO (fat mass and obesity-associated protein) within obesity; however, its functional role within different cell types is less understood. OBJECTIVES: We identify endothelial FTO as a previously unknown central regulator of both obesity-induced metabolic and vascular alterations. METHODS AND RESULTS: We generated endothelial Fto-deficient mice and analyzed the impact of obesity on those mice. While the loss of endothelial FTO did not influence the development of obesity and dyslipidemia, it protected mice from high-fat diet-induced glucose intolerance and insulin resistance by increasing AKT (protein kinase B) phosphorylation in endothelial cells and skeletal muscle. Furthermore, loss of endothelial FTO prevented the development of obesity-induced hypertension by preserving myogenic tone in resistance arteries. In Fto-deficient arteries, microarray analysis identified upregulation of L-Pgds with significant increases in prostaglandin D2 levels. Blockade of prostaglandin D2 synthesis inhibited the myogenic tone protection in resistance arteries of endothelial Fto-deficient mice on high-fat diet; conversely, direct addition of prostaglandin D2 rescued myogenic tone in high-fat diet-fed control mice. Myogenic tone was increased in obese human arteries with FTO inhibitors or prostaglandin D2 application. CONCLUSIONS: These data identify endothelial FTO as a previously unknown regulator in the development of obesity-induced metabolic and vascular changes, which is independent of its known function in regulation of obesity.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Endothelium, Vascular/metabolism , Obesity/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Arteries/metabolism , Arteries/pathology , Endothelium, Vascular/pathology , Humans , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Male , Mice , Muscle Tonus , Muscle, Skeletal/metabolism , Obesity/genetics , Obesity/pathology , Prostaglandin D2/metabolism , Proto-Oncogene Proteins c-akt/metabolism
12.
Physiol Rep ; 7(6): e14010, 2019 03.
Article in English | MEDLINE | ID: mdl-30916484

ABSTRACT

Hypertension is a major health concern in the developed world, and its prevalence increases with advancing age. The impact of hypertension on the function of the renal and cardiovascular systems is well studied; however, its influence on the brain regions important for cognition has garnered less attention. We utilized the Cyp1a1-Ren2 xenobiotic-inducible transgenic rat model to mimic both the age of onset and rate of induction of hypertension observed in humans. Male, 15-month-old transgenic rats were fed 0.15% indole-3-carbinol (I3C) chow to slowly induce renin-dependent hypertension over a 6-week period. Systolic blood pressure significantly increased, eventually reaching 200 mmHg by the end of the study period. In contrast, transgenic rats fed a control diet without I3C did not show significant changes in blood pressure (145 mmHg at the end of study). Hypertension was associated with cardiac, aortic, and renal hypertrophy as well as increased collagen deposition in the left ventricle and kidney of the I3C-treated rats. Additionally, rats with hypertension showed reduced savings from prior spatial memory training when tested on the hippocampus-dependent Morris swim task. Motor and sensory functions were found to be unaffected by induction of hypertension. Taken together, these data indicate a profound effect of hypertension not only on the cardiovascular-renal axis but also on brain systems critically important for learning and memory. Future use of this model and approach may empower a more accurate investigation of the influence of aging on the systems responsible for cardiovascular, renal, and neurological health.


Subject(s)
Behavior, Animal , Blood Pressure , Brain/physiopathology , Cytochrome P-450 CYP1A1/metabolism , Hypertension/physiopathology , Hypertension/psychology , Renin-Angiotensin System , Renin/metabolism , Spatial Learning , Animals , Blood Pressure/genetics , Cytochrome P-450 CYP1A1/genetics , Disease Models, Animal , Hypertension/chemically induced , Hypertension/genetics , Indoles , Locomotion , Male , Promoter Regions, Genetic , Rats, Inbred F344 , Rats, Transgenic , Renin/genetics , Renin-Angiotensin System/genetics , Time Factors
13.
Am J Hypertens ; 32(2): 123-134, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30380007

ABSTRACT

The mineralocorticoid receptor (MR) is indispensable for survival through its critical role in maintaining blood pressure in response to sodium scarcity or bleeding. Activation of MR by aldosterone in the kidney controls water and electrolyte homeostasis. This review summarizes recent advances in our understanding of MR function, specifically in vascular endothelial and smooth muscle cells. The evolving roles for vascular MR are summarized in the areas of (i) vascular tone regulation, (ii) thrombosis, (iii) inflammation, and (iv) vascular remodeling/fibrosis. Synthesis of the data supports the concept that vascular MR does not contribute substantially to basal homeostasis but rather, MR is poised to be activated when the vasculature is damaged to coordinate blood pressure maintenance and wound healing. Specifically, MR activation in the vascular wall promotes vasoconstriction, inflammation, and exuberant vascular remodeling with fibrosis. A teleological model is proposed in which these functions of vascular MR may have provided a critical evolutionary survival advantage in the face of mechanical vascular injury with bleeding. However, modern lifestyle is characterized by physical inactivity and high fat/high sodium diet resulting in diffuse vascular damage. Under these modern conditions, diffuse, persistent and unregulated activation of vascular MR contributes to post-reproductive cardiovascular disease in growing populations with hypertension, obesity, and advanced age.


Subject(s)
Cardiovascular Diseases/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hemodynamics , Life Style , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Mineralocorticoid/metabolism , Vascular Remodeling , Animals , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Diet, High-Fat , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Evolution, Molecular , Humans , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Risk Factors , Sedentary Behavior , Signal Transduction , Sodium, Dietary/adverse effects , Wound Healing
14.
J Clin Invest ; 128(11): 5073-5082, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30295646

ABSTRACT

Arteriolar endothelial cell-expressed (EC-expressed) α-globin binds endothelial NOS (eNOS) and degrades its enzymatic product, NO, via dioxygenation, thereby lessening the vasodilatory effects of NO on nearby vascular smooth muscle. Although this reaction potentially affects vascular physiology, the mechanisms that regulate α-globin expression and dioxygenase activity in ECs are unknown. Without ß-globin, α-globin is unstable and cytotoxic, particularly in its oxidized form, which is generated by dioxygenation and recycled via endogenous reductases. We show that the molecular chaperone α-hemoglobin-stabilizing protein (AHSP) promotes arteriolar α-globin expression in vivo and facilitates its reduction by eNOS. In Ahsp-/- mice, EC α-globin was decreased by 70%. Ahsp-/- and Hba1-/- mice exhibited similar evidence of increased vascular NO signaling, including arteriolar dilation, blunted α1-adrenergic vasoconstriction, and reduced blood pressure. Purified α-globin bound eNOS or AHSP, but not both together. In ECs in culture, eNOS or AHSP enhanced α-globin expression posttranscriptionally. However, only AHSP prevented oxidized α-globin precipitation in solution. Finally, eNOS reduced AHSP-bound α-globin approximately 6-fold faster than did the major erythrocyte hemoglobin reductases (cytochrome B5 reductase plus cytochrome B5). Our data support a model whereby redox-sensitive shuttling of EC α-globin between AHSP and eNOS regulates EC NO degradation and vascular tone.


Subject(s)
Endothelial Cells/metabolism , Models, Cardiovascular , Molecular Chaperones/metabolism , Muscle Contraction , Signal Transduction , alpha-Globins/metabolism , Animals , Arterioles , Endothelial Cells/cytology , Mice , Mice, Knockout , Molecular Chaperones/genetics , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , alpha-Globins/genetics
15.
Arterioscler Thromb Vasc Biol ; 38(1): 120-130, 2018 01.
Article in English | MEDLINE | ID: mdl-29122814

ABSTRACT

OBJECTIVE: In resistance arteries, endothelial cell (EC) extensions can make contact with smooth muscle cells, forming myoendothelial junction at holes in the internal elastic lamina (HIEL). At these HIEL, calcium signaling is tightly regulated. Because Calr (calreticulin) can buffer ≈50% of endoplasmic reticulum calcium and is expressed throughout IEL holes in small arteries, the only place where myoendothelial junctions form, we investigated the effect of EC-specific Calr deletion on calcium signaling and vascular function. APPROACH AND RESULTS: We found Calr expressed in nearly every IEL hole in third-order mesenteric arteries, but not other ER markers. Because of this, we generated an EC-specific, tamoxifen inducible, Calr knockout mouse (EC Calr Δ/Δ). Using this mouse, we tested third-order mesenteric arteries for changes in calcium events at HIEL and vascular reactivity after application of CCh (carbachol) or PE (phenylephrine). We found that arteries from EC Calr Δ/Δ mice stimulated with CCh had unchanged activity of calcium signals and vasodilation; however, the same arteries were unable to increase calcium events at HIEL in response to PE. This resulted in significantly increased vasoconstriction to PE, presumably because of inhibited negative feedback. In line with these observations, the EC Calr Δ/Δ had increased blood pressure. Comparison of ER calcium in arteries and use of an ER-specific GCaMP indicator in vitro revealed no observable difference in ER calcium with Calr knockout. Using selective detergent permeabilization of the artery and inhibition of Calr translocation, we found that the observed Calr at HIEL may not be within the ER. CONCLUSIONS: Our data suggest that Calr specifically at HIEL may act in a non-ER dependent manner to regulate arteriolar heterocellular communication and blood pressure.


Subject(s)
Blood Pressure , Calbindin 2/metabolism , Calcium Signaling , Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Mesenteric Arteries/metabolism , Myocytes, Smooth Muscle/metabolism , Paracrine Communication , Vasoconstriction , Animals , Blood Pressure/drug effects , Calbindin 2/deficiency , Calbindin 2/genetics , Calcium Signaling/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Humans , Intercellular Junctions/drug effects , Male , Mesenteric Arteries/drug effects , Mice, Inbred DBA , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Paracrine Communication/drug effects , Phenylephrine/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation
16.
J Vis Exp ; (127)2017 09 08.
Article in English | MEDLINE | ID: mdl-28930992

ABSTRACT

The myoendothelial junction (MEJ), a unique signaling microdomain in small diameter resistance arteries, exhibits localization of specific proteins and signaling processes that can control vascular tone and blood pressure. As it is a projection from either the endothelial or smooth muscle cell, and due to its small size (on average, an area of ~1 µm2), the MEJ is difficult to study in isolation. However, we have developed a cell culture model called the vascular cell co-culture (VCCC) that allows for in vitro MEJ formation, endothelial cell polarization, and dissection of signaling proteins and processes in the vascular wall of resistance arteries. The VCCC has a multitude of applications and can be adapted to suit different cell types. The model consists of two cell types grown on opposite sides of a filter with 0.4 µm pores in which the in vitro MEJs can form. Here we describe how to create the VCCC via plating of cells and isolation of endothelial, MEJ, and smooth muscle fractions, which can then be used for protein isolation or activity assays. The filter with intact cell layers can be fixed, embedded, and sectioned for immunofluorescent analysis. Importantly, many of the discoveries from this model have been confirmed using intact resistance arteries, underscoring its physiological relevance.


Subject(s)
Arteries/physiopathology , Cell Culture Techniques/methods , Endothelial Cells/cytology , Myocytes, Smooth Muscle/cytology , Coculture Techniques , Humans , Vascular Resistance
17.
Biochim Biophys Acta ; 1861(7): 671-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27106139

ABSTRACT

In resistance arteries, endothelial cells (EC) make contact with smooth muscle cells (SMC), forming myoendothelial junctions (MEJ). Endothelial nitric oxide synthase (eNOS) is present in the luminal side of the EC (apical EC) and the basal side of the EC (MEJ). To test if these eNOS pools acted in sync or separately, we co-cultured ECs and SMCs, then stimulated SMCs with phenylephrine (PE). Adrenergic activation causes inositol [1,4,5] triphosphate (IP3) to move from SMC to EC through gap junctions at the MEJ. PE increases MEJ eNOS phosphorylation (eNOS-P) at S1177, but not in EC. Conversely, we used bradykinin (BK) to increase EC calcium; this increased EC eNOS-P but did not affect MEJ eNOS-P. Inhibiting gap junctions abrogated the MEJ eNOS-P after PE, but had no effect on BK eNOS-P. Differential lipid composition between apical EC and MEJ may account for the compartmentalized eNOS-P response. Indeed, DAG and phosphatidylserine are both enriched in MEJ. These lipids are cofactors for PKC activity, which was significantly increased at the MEJ after PE. Because PKC activity also relies on endoplasmic reticulum (ER) calcium release, we used thapsigargin and xestospongin C, BAPTA, and PKC inhibitors, which caused significant decreases in MEJ eNOS-P after PE. Functionally, BK inhibited leukocyte adhesion and PE caused an increase in SMC cGMP. We hypothesize that local lipid composition of the MEJ primes PKC and eNOS-P for stimulation by PE, allowing for compartmentalized function of eNOS in the blood vessel wall.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Endothelial Cells/enzymology , Gap Junctions/chemistry , Myocytes, Smooth Muscle/enzymology , Nitric Oxide Synthase Type III/metabolism , Biological Transport , Bradykinin/pharmacology , Calcium Signaling , Cell Communication/drug effects , Coculture Techniques , Cyclic GMP/metabolism , Diglycerides/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Endoplasmic Reticulum/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gap Junctions/drug effects , Gap Junctions/metabolism , Gene Expression Regulation , Humans , Inositol 1,4,5-Trisphosphate , Macrocyclic Compounds/pharmacology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Nitric Oxide Synthase Type III/classification , Nitric Oxide Synthase Type III/genetics , Oxazoles/pharmacology , Phenylephrine/pharmacology , Phosphatidylserines/metabolism , Phosphorylation , Primary Cell Culture , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Thapsigargin/pharmacology
18.
Hypertens Res ; 39(1): 8-18, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26490086

ABSTRACT

We have previously demonstrated that angiotensin-converting enzyme (ACE) inhibition with enalapril produces persistent effects that protect against future nitric oxide synthase (NOS) inhibitor (L-arginine methyl ester, L-NAME)-induced cardiac dysfunction and outer wall collagen deposition in spontaneously hypertensive rats (SHR). In the present study, we dissect the cytokine/chemokine release profile during NOS inhibition, its correlation to pathological cardiac remodeling and the impact of transient ACE inhibition on these effects. Adult male SHR were treated with enalapril (E+L) or tap water (C+L) for 2 weeks followed by a 2-week washout period. Rats were then subjected to 0, 3, 7 or 10 days of L-NAME treatment. The temporal response to NOS inhibition was evaluated by measuring arterial pressure, cardiac remodeling and cytokine/chemokine levels. L-NAME equivalently increased blood pressure and myocardial and vascular injury in C+L and E+L rats. However, pulse pressure (PP) was only transiently altered in C+L rats. The levels of several inflammatory mediators were increased during L-NAME treatment. However, interleukin-6 (IL-6) and IL-10 and monocyte chemoattractant protein-1 were uniquely increased in C+L hearts; whereas IL-4 and fractalkine were only elevated in E+L hearts. By days 7 and 10 of L-NAME treatment, there was a significant increase in the cardiac density of macrophages and proliferating cells, respectively only in C+L rats. Although myocardial injury was similar in both treatment groups, PP was not changed and there was a distinct cardiac chemokine/cytokine signature in rats previously treated with enalapril that may be related to the lack of proliferative response and macrophage infiltration in these hearts.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Myocarditis/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Arterial Pressure/drug effects , Cell Proliferation/drug effects , Chemokine CCL2/blood , Chemokines , Cytokines/metabolism , Disease Progression , Enalapril/pharmacology , Interleukin-10/blood , Interleukin-6/blood , Macrophages/drug effects , Male , Myocarditis/chemically induced , Myocarditis/prevention & control , Rats , Rats, Inbred SHR
19.
Pharmacol Rev ; 66(2): 513-69, 2014.
Article in English | MEDLINE | ID: mdl-24671377

ABSTRACT

It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.


Subject(s)
Blood Vessels , Cell Communication/physiology , Cell Membrane/metabolism , Endothelium, Vascular/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Microdomains/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/metabolism , Endothelium, Vascular/cytology , Gap Junctions/metabolism , Humans , Ion Channels/metabolism , Paracrine Communication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...