Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Respir Crit Care Med ; 206(9): 1128-1139, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35771569

ABSTRACT

Rationale: Treatment options for idiopathic pulmonary fibrosis (IPF) are limited. Objectives: To evaluate the efficacy and safety of BG00011, an anti-αvß6 IgG1 monoclonal antibody, in the treatment of patients with IPF. Methods: In a phase IIb randomized, double-blind, placebo-controlled trial, patients with IPF (FVC ⩾50% predicted, on or off background therapy) were randomized 1:1 to once-weekly subcutaneous BG00011 56 mg or placebo. The primary endpoint was FVC change from baseline at Week 52. Because of early trial termination (imbalance in adverse events and lack of clinical benefit), endpoints were evaluated at Week 26 as an exploratory analysis. Measurements and Main Results: One hundred six patients were randomized and received at least one dose of BG00011 (n = 54) or placebo (n = 52). At Week 26, there was no significant difference in FVC change from baseline between patients who received BG00011 (n = 20) or placebo (n = 23), least squares mean (SE) -0.097 L (0.0600) versus -0.056 L (0.0593), respectively (P = 0.268). However, after Week 26, patients in the BG00011 group showed a worsening trend. Eight (44.4%) of 18 who received BG00011 and 4 (18.2%) of 22 who received placebo showed worsening of fibrosis on high-resolution computed tomography at the end of treatment. IPF exacerbation/or progression was reported in 13 patients (all in the BG00011 group). Serious adverse events occurred more frequently in BG00011 patients, including four deaths. Conclusions: The results do not support the continued clinical development of BG00011. Further research is warranted to identify new treatment strategies that modify inflammatory and fibrotic pathways in IPF. Clinical trial registered with www.clinicaltrials.gov (NCT03573505).


Subject(s)
Idiopathic Pulmonary Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Antibodies, Monoclonal/therapeutic use , Treatment Outcome , Double-Blind Method , Immunoglobulin G
2.
Hypertension ; 71(4): 609-621, 2018 04.
Article in English | MEDLINE | ID: mdl-29463624

ABSTRACT

Stiffening of the vasculature with aging is a strong predictor of adverse cardiovascular events, independent of all other risk factors including blood pressure, yet no therapies target this process. MRs (mineralocorticoid receptors) in smooth muscle cells (SMCs) have been implicated in the regulation of vascular fibrosis but have not been explored in vascular aging. Comparing SMC-MR-deleted male mice to MR-intact littermates at 3, 12, and 18 months of age, we demonstrated that aging-associated vascular stiffening and fibrosis are mitigated by MR deletion in SMCs. Progression of cardiac stiffness and fibrosis and the decline in exercise capacity with aging were also mitigated by MR deletion in SMC. Vascular gene expression profiling analysis revealed that MR deletion in SMC is associated with recruitment of a distinct antifibrotic vascular gene expression program with aging. Moreover, long-term pharmacological inhibition of MR in aged mice prevented the progression of vascular fibrosis and stiffness and induced a similar antifibrotic vascular gene program. Finally, in a small trial in elderly male humans, short-term MR antagonism produced an antifibrotic signature of circulating biomarkers similar to that observed in the vasculature of SMC-MR-deleted mice. These findings suggest that SMC-MR contributes to vascular stiffening with aging and is a potential therapeutic target to prevent the progression of aging-associated vascular fibrosis and stiffness.


Subject(s)
Cellular Senescence , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Receptors, Mineralocorticoid , Spironolactone , Vascular Stiffness , Aged , Animals , Cellular Senescence/drug effects , Cellular Senescence/physiology , Disease Progression , Exercise Tolerance/physiology , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Gene Expression/drug effects , Gene Expression Profiling , Humans , Male , Mice , Mineralocorticoid Receptor Antagonists/metabolism , Mineralocorticoid Receptor Antagonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Spironolactone/metabolism , Spironolactone/pharmacology , Treatment Outcome , Vascular Stiffness/drug effects , Vascular Stiffness/physiology
3.
J Am Soc Nephrol ; 28(6): 1741-1752, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28062569

ABSTRACT

Ischemia-reperfusion injury (IRI) is a leading cause of AKI. This common clinical complication lacks effective therapies and can lead to the development of CKD. The αvß5 integrin may have an important role in acute injury, including septic shock and acute lung injury. To examine its function in AKI, we utilized a specific function-blocking antibody to inhibit αvß5 in a rat model of renal IRI. Pretreatment with this anti-αvß5 antibody significantly reduced serum creatinine levels, diminished renal damage detected by histopathologic evaluation, and decreased levels of injury biomarkers. Notably, therapeutic treatment with the αvß5 antibody 8 hours after IRI also provided protection from injury. Global gene expression profiling of post-ischemic kidneys showed that αvß5 inhibition affected established injury markers and induced pathway alterations previously shown to be protective. Intravital imaging of post-ischemic kidneys revealed reduced vascular leak with αvß5 antibody treatment. Immunostaining for αvß5 in the kidney detected evident expression in perivascular cells, with negligible expression in the endothelium. Studies in a three-dimensional microfluidics system identified a pericyte-dependent role for αvß5 in modulating vascular leak. Additional studies showed αvß5 functions in the adhesion and migration of kidney pericytes in vitro Initial studies monitoring renal blood flow after IRI did not find significant effects with αvß5 inhibition; however, future studies should explore the contribution of vasomotor effects. These studies identify a role for αvß5 in modulating injury-induced renal vascular leak, possibly through effects on pericyte adhesion and migration, and reveal αvß5 inhibition as a promising therapeutic strategy for AKI.


Subject(s)
Capillary Permeability/drug effects , Kidney/blood supply , Receptors, Vitronectin/antagonists & inhibitors , Reperfusion Injury/prevention & control , Animals , Male , Rats , Rats, Sprague-Dawley
4.
JCI Insight ; 1(14): e88942, 2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27683672

ABSTRACT

Hypertension is nearly universal yet poorly controlled in the elderly despite proven benefits of intensive treatment. Mice lacking mineralocorticoid receptors in smooth muscle cells (SMC-MR-KO) are protected from rising blood pressure (BP) with aging, despite normal renal function. Vasoconstriction is attenuated in aged SMC-MR-KO mice, thus they were used to explore vascular mechanisms that may contribute to hypertension with aging. MicroRNA (miR) profiling identified miR-155 as the most down-regulated miR with vascular aging in MR-intact but not SMC-MR-KO mice. The aging-associated decrease in miR-155 in mesenteric resistance vessels was associated with increased mRNA abundance of MR and of predicted miR-155 targets Cav1.2 (L-type calcium channel (LTCC) subunit) and angiotensin type-1 receptor (AgtR1). SMC-MR-KO mice lacked these aging-associated vascular gene expression changes. In HEK293 cells, MR repressed miR-155 promoter activity. In cultured SMCs, miR-155 decreased Cav1.2 and AgtR1 mRNA. Compared to MR-intact littermates, aged SMC-MR-KO mice had decreased systolic BP, myogenic tone, SMC LTCC current, mesenteric vessel calcium influx, LTCC-induced vasoconstriction and angiotensin II-induced vasoconstriction and oxidative stress. Restoration of miR-155 specifically in SMCs of aged MR-intact mice decreased Cav1.2 and AgtR1 mRNA and attenuated LTCC-mediated and angiotensin II-induced vasoconstriction and oxidative stress. Finally, in a trial of MR blockade in elderly humans, changes in serum miR-155 predicted the BP treatment response. Thus, SMC-MR regulation of miR-155, Cav1.2 and AgtR1 impacts vasoconstriction with aging. This novel mechanism identifies potential new treatment strategies and biomarkers to improve and individualize antihypertensive therapy in the elderly.

5.
J Clin Invest ; 126(7): 2561-74, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27270170

ABSTRACT

Preeclampsia is a hypertensive disorder of pregnancy in which patients develop profound sensitivity to vasopressors, such as angiotensin II, and is associated with substantial morbidity for the mother and fetus. Enhanced vasoconstrictor sensitivity and elevations in soluble fms-like tyrosine kinase 1 (sFLT1), a circulating antiangiogenic protein, precede clinical signs and symptoms of preeclampsia. Here, we report that overexpression of sFlt1 in pregnant mice induced angiotensin II sensitivity and hypertension by impairing endothelial nitric oxide synthase (eNOS) phosphorylation and promoting oxidative stress in the vasculature. Administration of the NOS inhibitor l-NAME to pregnant mice recapitulated the angiotensin sensitivity and oxidative stress observed with sFlt1 overexpression. Sildenafil, an FDA-approved phosphodiesterase 5 inhibitor that enhances NO signaling, reversed sFlt1-induced hypertension and angiotensin II sensitivity in the preeclampsia mouse model. Sildenafil treatment also improved uterine blood flow, decreased uterine vascular resistance, and improved fetal weights in comparison with untreated sFlt1-expressing mice. Finally, sFLT1 protein expression inversely correlated with reductions in eNOS phosphorylation in placental tissue of human preeclampsia patients. These data support the concept that endothelial dysfunction due to high circulating sFLT1 may be the primary event leading to enhanced vasoconstrictor sensitivity that is characteristic of preeclampsia and suggest that targeting sFLT1-induced pathways may be an avenue for treating preeclampsia and improving fetal outcomes.


Subject(s)
Angiotensin II/metabolism , Nitric Oxide Synthase Type III/metabolism , Pre-Eclampsia/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Angiotensins/metabolism , Animals , Blood Pressure , Disease Models, Animal , Female , Humans , Male , Mice , NG-Nitroarginine Methyl Ester/chemistry , Oxidative Stress , Phosphorylation , Placenta/metabolism , Pregnancy , Pregnancy, Animal , Signal Transduction , Sildenafil Citrate/therapeutic use , Treatment Outcome
6.
Hypertension ; 65(4): 863-70, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712723

ABSTRACT

Cardiovascular disease (CVD) remains the leading killer of women in developed nations. One sex-specific risk factor is preeclampsia, a syndrome of hypertension and proteinuria that complicates 5% of pregnancies. Although preeclampsia resolves after delivery, exposed women are at increased long-term risk of premature CVD and mortality. Pre-existing CVD risk factors are associated with increased risk of developing preeclampsia but whether preeclampsia merely uncovers risk or contributes directly to future CVD remains a critical unanswered question. A mouse preeclampsia model was used to test the hypothesis that preeclampsia causes an enhanced vascular response to future vessel injury. A preeclampsia-like state was induced in pregnant CD1 mice by overexpressing soluble fms-like tyrosine kinase-1, a circulating antiangiogenic protein that induces hypertension and glomerular disease resembling human preeclampsia. Two months postpartum, soluble fms-like tyrosine kinase-1 levels and blood pressure normalized and cardiac size and function by echocardiography and renal histology were indistinguishable in preeclampsia-exposed compared with control mice. Mice were then challenged with unilateral carotid injury. Preeclampsia-exposed mice had significantly enhanced vascular remodeling with increased vascular smooth muscle cell proliferation (180% increase; P<0.01) and vessel fibrosis (216% increase; P<0.001) compared with control pregnancy. In the contralateral uninjured vessel, there was no difference in remodeling after exposure to preeclampsia. These data support a new model in which vessels exposed to preeclampsia retain a persistently enhanced vascular response to injury despite resolution of preeclampsia after delivery. This new paradigm may contribute to the substantially increased risk of CVD in woman exposed to preeclampsia.


Subject(s)
Carotid Arteries/pathology , Carotid Artery Diseases/etiology , Pre-Eclampsia/physiopathology , Pregnancy, Animal , Recovery of Function , Vascular Remodeling , Animals , Blood Pressure/physiology , Carotid Arteries/diagnostic imaging , Carotid Arteries/physiopathology , Carotid Artery Diseases/pathology , Carotid Artery Diseases/physiopathology , Disease Models, Animal , Female , Mice , Postpartum Period , Pre-Eclampsia/diagnostic imaging , Pregnancy , Ultrasonography
7.
Endocrinology ; 155(11): 4461-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25051445

ABSTRACT

The steroid hormone aldosterone (aldo) contributes to cardiovascular disease in animal models and in humans. Aldo activates the mineralocorticoid receptor (MR), a hormone-activated transcription factor, and indeed, pharmacological MR inhibition improves cardiovascular outcomes. Because the incidence of cardiovascular disease is lower in premenopausal women, we hypothesized that estrogen (E2) signaling through the estrogen receptor (ER) may protect the vasculature by inhibiting the detrimental effects of aldo signaling through the MR. We demonstrate that E2-activated ER inhibits MR-mediated gene transcription from the mouse mammary tumor virus reporter in human embryonic kidney-293 cells. In contrast, aldo-activated MR does not affect ER-mediated gene transcription. The ERα N terminus (amino acids 1-253) containing part of the DNA-binding domain is sufficient to inhibit MR genomic function, although point mutations reveal that DNA binding, ligand-independent activation, and rapid nongenomic ERα signaling are not required for this effect. Furthermore, ERα and MR are part of a complex in cell lysates, with amino acids 1-233 of the ERα N terminus being sufficient to complex with the MR. Overall, the ability of ERα to inhibit MR-mediated gene transcription correlates with the ability of ERα segments to both localize to the nucleus and complex with the MR. In cultured vascular endothelial cells expressing ERα, E2 inhibits aldo induction of the vascular MR target gene intercellular adhesion molecule-1 (ICAM-1). ICAM-1 induction by endothelial MR is known to promote vascular inflammation that could contribute to the mechanism of aldo-induced atherosclerosis. E2 also inhibits aldo induction of ICAM-1 protein and prevents aldo-enhanced leukocyte adhesion to endothelial cells. These studies support a new model in which E2-activated ER in endothelial cells forms a complex with MR in the nucleus to modulate MR regulation of the proinflammatory gene ICAM-1. Estrogen inhibition of MR regulation of genes that contribute to cardiovascular disease may be a new mechanism by which premenopausal women are protected from cardiovascular disease.


Subject(s)
Estrogens/pharmacology , Gene Expression Regulation , Mineralocorticoid Receptor Antagonists/pharmacology , Receptors, Estrogen/physiology , Receptors, Mineralocorticoid/physiology , Animals , Cells, Cultured , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Protein Binding , Protein Structure, Tertiary , Receptors, Estrogen/chemistry , Receptors, Mineralocorticoid/chemistry , U937 Cells , Xenopus
8.
Arterioscler Thromb Vasc Biol ; 34(2): 355-64, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24311380

ABSTRACT

OBJECTIVE: Vascular remodeling occurs after endothelial injury, resulting in smooth muscle cell (SMC) proliferation and vascular fibrosis. We previously demonstrated that the blood pressure-regulating hormone aldosterone enhances vascular remodeling in mice at sites of endothelial injury in a placental growth factor-dependent manner. We now test the hypothesis that SMC mineralocorticoid receptors (MRs) directly mediate the remodeling effects of aldosterone and further explore the mechanism. APPROACH AND RESULTS: A wire-induced carotid injury model was performed in wild-type mice and mice with inducible SMC-specific deletion of the MR. Aldosterone did not affect re-endothelialization after injury in wild-type mice. Deletion of SMC-MR prevented the 79% increase in SMC proliferation induced by aldosterone after injury in MR-Intact littermates. Moreover, both injury-induced and aldosterone-enhanced vascular fibrosis were attenuated in SMC-specific MR knockout mice. Further exploration of the mechanism revealed that aldosterone-induced vascular remodeling is prevented by in vivo blockade of the placental growth factor-specific receptor, type 1 vascular endothelial growth factor receptor (VEGFR1), the receptor for placental growth factor. Immunohistochemistry of carotid vessels shows that the induction of VEGFR1 expression in SMC after vascular injury is attenuated by 72% in SMC-specific MR knockout mice. Moreover, aldosterone induction of vascular placental growth factor mRNA expression and protein release are also prevented in vessels lacking SMC-MR. CONCLUSIONS: These studies reveal that SMC-MR is necessary for aldosterone-induced vascular remodeling independent of renal effects on blood pressure. SMC-MR contributes to induction of SMC VEGFR1 in the area of vascular injury and to aldosterone-enhanced vascular placental growth factor expression and hence the detrimental effects of aldosterone are prevented by VEGFR1 blockade. This study supports exploring MR antagonists and VEGFR1 blockade to prevent pathological vascular remodeling induced by aldosterone.


Subject(s)
Aldosterone/pharmacology , Carotid Artery Injuries/metabolism , Cell Proliferation/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, Mineralocorticoid/agonists , Animals , Antibodies/pharmacology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrosis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Mineralocorticoid/deficiency , Receptors, Mineralocorticoid/genetics , Time Factors , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism
9.
J Am Soc Nephrol ; 25(4): 717-25, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24335973

ABSTRACT

Soluble fms-like tyrosine kinase 1 (sFlt1), a circulating antiangiogenic protein, is elevated in kidney diseases and contributes to the development of preeclampsia. Hydrogen sulfide is a vasorelaxant and proangiogenic gas with therapeutic potential in several diseases. Therefore, we evaluated the potential therapeutic effect and mechanisms of action of hydrogen sulfide in an animal model of sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis created by adenovirus-mediated overexpression of sFlt1 in Sprague-Dawley rats. We injected sFlt1-overexpressing animals intraperitoneally with the hydrogen sulfide-donor sodium hydrosulfide (NaHS) (50 µmol/kg, twice daily) or vehicle (n=7 per group). Treatment with NaHS for 8 days significantly reduced sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis. Measurement of plasma protein concentrations with ELISA revealed a reduction of free plasma sFlt1 and an increase of free plasma vascular endothelial growth factor (VEGF) after treatment with NaHS. Renal VEGF-A mRNA expression increased significantly with NaHS treatment. In vitro, NaHS was proangiogenic in an endothelial tube assay and attenuated the antiangiogenic effects of sFlt1. Stimulation of podocytes with NaHS resulted in both short-term VEGF release (120 minutes) and upregulation of VEGF-A mRNA levels (24 hours). Furthermore, pretreatment of mesenteric vessels with a VEGF receptor 2-neutralizing antibody significantly attenuated NaHS-induced vasodilation. These results suggest that hydrogen sulfide ameliorates sFlt1-induced hypertension, proteinuria, and glomerular endotheliosis in rats by increasing VEGF expression. Further studies are warranted to evaluate the role of hydrogen sulfide as a novel therapeutic agent for vascular disorders such as preeclampsia.


Subject(s)
Hydrogen Sulfide/pharmacology , Hypertension/drug therapy , Proteinuria/drug therapy , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-1/physiology , Animals , Female , Kidney Glomerulus/drug effects , Podocytes/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vasodilation/drug effects
10.
Pflugers Arch ; 465(12): 1661-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23636772

ABSTRACT

The mineralocorticoid receptor (MR), a member of the steroid receptor family, regulates blood pressure by mediating the effects of the hormone aldosterone on renal sodium handling. In recent years, it has become clear that MR is expressed in vascular smooth muscle cells (SMCs), and interest has grown in understanding the direct role of SMC MR in regulating vascular function. This interest stems from multiple clinical studies where MR inhibitor treatment reduced the incidence of cardiovascular events and mortality. This review summarizes the most recent advances in our understanding of SMC MR in regulating normal vascular function and in promoting vascular disease. Many new studies suggest a role for SMC MR activation in stimulating vascular contraction and contributing to vessel inflammation, fibrosis, and remodeling. These detrimental vascular effects of MR activation appear to be independent of changes in blood pressure and are synergistic with the presence of endothelial dysfunction or damage. Thus, in humans with underlying cardiovascular disease or cardiovascular risk factors, SMC MR activation may promote hypertension, atherosclerosis, and vascular aging. Further exploration of the molecular mechanisms for the effects of SMC MR activation has the potential to identify novel therapeutic targets to prevent or treat common cardiovascular disorders.


Subject(s)
Aldosterone/physiology , Cardiovascular Diseases/physiopathology , Muscle, Smooth, Vascular/physiology , Receptors, Mineralocorticoid/physiology , Aging , Angiotensin II/physiology , Animals , Atherosclerosis/etiology , Blood Pressure/drug effects , Cardiovascular Diseases/pathology , Cardiovascular Physiological Phenomena , Carotid Arteries/drug effects , Carotid Arteries/pathology , Fibrosis , Gene Expression Regulation/drug effects , Humans , Hypertension/etiology , Mice , Muscle, Smooth, Vascular/metabolism , Placenta Growth Factor , Pregnancy Proteins/biosynthesis , Signal Transduction/physiology
11.
Curr Atheroscler Rep ; 15(7): 340, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23719923

ABSTRACT

The mineralocorticoid receptor (MR), a steroid-hormone-activated transcription factor, plays a substantial role in cardiovascular diseases. MR antagonists (MRAs) have long been appreciated as effective treatments for heart failure and hypertension; however, recent research suggests that additional patient populations may also benefit from MRA therapy. Experimental evidence demonstrates that in addition to its classic role in the regulating sodium handling in the kidney, functional MR is expressed in the blood vessels and contributes to hypertension, vascular inflammation and remodeling, and atherogenesis. MR activation drives pathological phenotypes in smooth muscle cells, endothelial cells, and inflammatory cells, whereas MRAs inhibit these effects. Collectively, these studies demonstrate a new role for extrarenal MR in cardiovascular disease. This review summarizes these new lines of evidence and how they contribute to the mechanisms of atherosclerosis, pulmonary and systemic hypertension, and vein graft failure, and describes new patient populations that may benefit from MRA therapy.


Subject(s)
Atherosclerosis/physiopathology , Receptors, Mineralocorticoid/physiology , Vascular Diseases/drug therapy , Vascular Diseases/physiopathology , Aldosterone/therapeutic use , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Disease Progression , Endothelium, Vascular/physiopathology , Humans , Hypertension/drug therapy , Hypertension/physiopathology , Mineralocorticoid Receptor Antagonists/therapeutic use , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Veins/transplantation
12.
Clin Exp Pharmacol Physiol ; 40(12): 902-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23710823

ABSTRACT

Hypertension is an extremely prevalent cardiovascular risk factor and current antihypertensive therapies do not adequately treat hypertension in many affected individuals. Thus, a better understanding of mechanisms of hypertension could lead to novel therapies. Mineralocorticoid receptors (MR) are known to regulate blood pressure by responding to aldosterone in the kidney to regulate sodium retention. Recent evidence supports a direct contribution of the vasculature to control of BP and suggests the possibility that MR antagonists may also lower blood pressure by acting on extrarenal MR. This review summarizes existing research considering the role of the vascular MR in regulating vasoreactivity and blood pressure. Multiple studies indicate a role for vascular MR in modulating vasoconstriction and vasorelaxation. Activation of MR in vascular endothelial and smooth muscle cells leads to increased reactive oxygen species production and decreased availability of nitric oxide, important regulators of vascular reactivity. Transgenic mouse models, including an endothelial MR overexpressing mouse and a smooth muscle cell-specific MR-knockout mouse, support a direct role for vascular MR in control of blood pressure. This new evidence demonstrating that vascular MR directly contribute to control of vasoreactivity and blood pressure supports vascular MR and the pathways they control as novel therapeutic targets to treat hypertension.


Subject(s)
Blood Pressure , Endothelium, Vascular/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Mineralocorticoid/metabolism , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Endothelium, Vascular/drug effects , Humans , Hypertension/drug therapy , Mineralocorticoid Receptor Antagonists/therapeutic use , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Vasodilation/drug effects
13.
Nat Med ; 18(9): 1429-33, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22922412

ABSTRACT

Hypertension is a cardiovascular risk factor present in over two-thirds of people over age 60 in North America; elevated blood pressure correlates with increased risk of heart attack, stroke and progression to heart and kidney failure. Current therapies are insufficient to control blood pressure in almost half of these patients. The mineralocorticoid receptor (MR), acting in the kidney, is known to regulate blood pressure through aldosterone binding and stimulation of sodium retention. However, recent studies support the concept that the MR also has extrarenal actions and that defects in sodium handling alone do not fully explain the development of hypertension and associated cardiovascular mortality. We and others have identified functional MR in human vascular smooth muscle cells (SMCs), suggesting that vascular MR might directly regulate blood pressure. Here we show that mice with SMC-specific deficiency of the MR have decreased blood pressure as they age without defects in renal sodium handling or vascular structure. Aged mice lacking MR in SMCs (SMC-MR) have reduced vascular myogenic tone, agonist-dependent contraction and expression and activity of L-type calcium channels. Moreover, SMC-MR contributes to angiotensin II­induced vascular oxidative stress, vascular contraction and hypertension. This study identifies a new role for vascular MR in blood pressure control and in vascular aging and supports the emerging hypothesis that vascular tone contributes directly to systemic blood pressure.


Subject(s)
Aging/physiology , Blood Pressure/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Receptors, Mineralocorticoid/metabolism , Animals , Calcium Channels, L-Type/metabolism , DNA Primers/genetics , Galactosides , Heart/anatomy & histology , Indoles , Mice , Muscle Contraction/physiology , Organ Size/physiology , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Receptors, Mineralocorticoid/deficiency , Sodium/metabolism
14.
Mol Cell Endocrinol ; 350(2): 256-65, 2012 Mar 24.
Article in English | MEDLINE | ID: mdl-21723914

ABSTRACT

The mineralocorticoid receptor (MR), a member of the steroid receptor family, regulates blood pressure by mediating the effects of the hormone aldosterone (Aldo) on renal sodium handling. Over the past decade, it has become clear that MR is expressed in the cardiovascular system and interest has grown in understanding the direct role of the MR in regulating vascular function and contributing to cardiovascular disease. This interest stems from multiple clinical studies in which drugs that decrease MR activation also reduce the incidence of heart attacks, strokes, and mortality out of proportion to modest changes in systemic blood pressure. The presence of functional mineralocorticoid receptors in vascular smooth muscle and endothelial cells is now well established and, while still controversial, data supports the vasculature as an Aldo-responsive tissue. This review summarizes recent advances in our understanding of the role of vascular MR in regulating normal vascular function and in promoting vascular disease. In vitro data, in vivo animal studies, and human data are reviewed suggesting a role for MR-activation in promoting vascular oxidative stress, inhibiting vascular relaxation, and contributing to vessel inflammation, fibrosis, and remodeling. These detrimental vascular effects of MR activation appear to be independent of changes in blood pressure and are synergistic with the presence of endothelial dysfunction or damage. Thus, in humans with underlying cardiovascular disease or cardiovascular risk factors, vascular MR activation may promote vascular aging and atherosclerosis thereby contributing to the pathophysiology of heart attack, stroke, and possibly even hypertension. Further exploration of the molecular mechanisms for the detrimental vascular effects of MR activation has the potential to identify novel therapeutic targets to prevent or treat common cardiovascular disorders.


Subject(s)
Blood Vessels/physiology , Receptors, Mineralocorticoid/physiology , Vascular Diseases/genetics , Animals , Blood Vessels/metabolism , Cardiovascular Physiological Phenomena , Cardiovascular System/metabolism , Humans , Inflammation/genetics , Models, Biological , Oxidative Stress/genetics , Oxidative Stress/physiology , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Vascular Diseases/metabolism
15.
J Exp Neurosci ; 2010(4): 17-33, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20740047

ABSTRACT

It is well-established that neurons in the adult mammalian central nervous system (CNS) are terminally differentiated and, if injured, will be unable to regenerate their connections. In contrast to mammals, zebrafish and other teleosts display a robust neuroregenerative response. Following optic nerve crush (ONX), retinal ganglion cells (RGC) regrow their axons to synapse with topographically correct targets in the optic tectum, such that vision is restored in approximately 21 days. What accounts for these differences between teleostean and mammalian responses to neural injury is not fully understood. A time course analysis of global gene expression patterns in the zebrafish eye after ONX can help to elucidate cellular and molecular mechanisms that contribute to a successful neuroregeneration. To define different phases of regeneration after ONX, alpha tubulin 1 (tuba1) and growth-associated protein 43 (gap43), markers previously shown to correspond to morphophological events, were measured by real time quantitative PCR (qPCR). Microarray analysis was then performed at defined intervals (6 hours, 1, 4, 12, and 21 days) post-ONX and compared to SHAM. Results show that optic nerve damage induces multiple, phase-related transcriptional programs, with the maximum number of genes changed and highest fold-change occurring at 4 days. Several functional groups affected by optic nerve regeneration, including cell adhesion, apoptosis, cell cycle, energy metabolism, ion channel activity, and calcium signaling, were identified. Utilizing the whole eye allowed us to identify signaling contributions from the vitreous, immune and glial cells as well as the neural cells of the retina. Comparisons between our dataset and transcriptional profiles from other models of regeneration in zebrafish retina, heart and fin revealed a subset of commonly regulated transcripts, indicating shared mechanisms in different regenerating tissues. Knowledge of gene expression patterns in all components of the eye in a model of successful regeneration provides an entry point for functional analyses, and will help in devising hypotheses for testing normal and toxic regulatory factors.

16.
BMC Mol Biol ; 9: 102, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19014500

ABSTRACT

BACKGROUND: Research using the zebrafish model has experienced a rapid growth in recent years. Although real-time reverse transcription PCR (QPCR), normalized to an internal reference ("housekeeping") gene, is a frequently used method for quantifying gene expression changes in zebrafish, many commonly used housekeeping genes are known to vary with experimental conditions. To identify housekeeping genes that are stably expressed under different experimental conditions, and thus suitable as normalizers for QPCR in zebrafish, the present study evaluated the expression of eight commonly used housekeeping genes as a function of stage and hormone/toxicant exposure during development, and by tissue type and sex in adult fish. RESULTS: QPCR analysis was used to quantify mRNA levels of bactin1, tubulin alpha 1(tuba1), glyceraldehyde-3-phosphate dehydrogenase (gapdh), glucose-6-phosphate dehydrogenase (g6pd), TATA-box binding protein (tbp), beta-2-microglobulin (b2m), elongation factor 1 alpha (elfa), and 18s ribosomal RNA (18s) during development (2 - 120 hr postfertilization, hpf); in different tissue types (brain, eye, liver, heart, muscle, gonads) of adult males and females; and after treatment of embryos/larvae (24 - 96 hpf) with commonly used vehicles for administration and agents that represent known environmental endocrine disruptors. All genes were found to have some degree of variability under the conditions tested here. Rank ordering of expression stability using geNorm analysis identified 18s, b2m, and elfa as most stable during development and across tissue types, while gapdh, tuba1, and tpb were the most variable. Following chemical treatment, tuba1, bactin1, and elfa were the most stably expressed whereas tbp, 18s, and b2m were the least stable. Data also revealed sex differences that are gene- and tissue-specific, and treatment effects that are gene-, vehicle- and ligand-specific. When the accuracy of QPCR analysis was tested using different reference genes to measure suppression of cyp19a1b by an estrogen receptor antagonist and induction of cyp1a by an arylhydrocarbon receptor agonist, the direction and magnitude of effects with stable and unstable genes differed. CONCLUSION: This study provides data that can be expected to aid zebrafish researchers in their initial choice of housekeeping genes for future studies, but underlines the importance of further validating housekeeping genes for each new experimental paradigm and fish species.


Subject(s)
Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Embryo, Nonmammalian/metabolism , Female , Gene Expression , Gene Expression Profiling , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...