Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
PLoS One ; 18(3): e0255709, 2023.
Article in English | MEDLINE | ID: mdl-36940215

ABSTRACT

Glucocorticoids inhibit angiogenesis by activating the glucocorticoid receptor. Inhibition of the glucocorticoid-activating enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) reduces tissue-specific glucocorticoid action and promotes angiogenesis in murine models of myocardial infarction. Angiogenesis is important in the growth of some solid tumours. This study used murine models of squamous cell carcinoma (SCC) and pancreatic ductal adenocarcinoma (PDAC) to test the hypothesis that 11ß-HSD1 inhibition promotes angiogenesis and subsequent tumour growth. SCC or PDAC cells were injected into female FVB/N or C57BL6/J mice fed either standard diet, or diet containing the 11ß-HSD1 inhibitor UE2316. SCC tumours grew more rapidly in UE2316-treated mice, reaching a larger (P<0.01) final volume (0.158 ± 0.037 cm3) than in control mice (0.051 ± 0.007 cm3). However, PDAC tumour growth was unaffected. Immunofluorescent analysis of SCC tumours did not show differences in vessel density (CD31/alpha-smooth muscle actin) or cell proliferation (Ki67) after 11ß-HSD1 inhibition, and immunohistochemistry of SCC tumours did not show changes in inflammatory cell (CD3- or F4/80-positive) infiltration. In culture, the growth/viability (assessed by live cell imaging) of SCC cells was not affected by UE2316 or corticosterone. Second Harmonic Generation microscopy showed that UE2316 reduced Type I collagen (P<0.001), whilst RNA-sequencing revealed that multiple factors involved in the innate immune/inflammatory response were reduced in UE2316-treated SCC tumours. 11ß-HSD1 inhibition increases SCC tumour growth, likely via suppression of inflammatory/immune cell signalling and extracellular matrix deposition, but does not promote tumour angiogenesis or growth of all solid tumours.


Subject(s)
Glucocorticoids , Neoplasms , Mice , Female , Animals , Glucocorticoids/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Inflammation , Neovascularization, Pathologic , Fibrosis
2.
Front Cardiovasc Med ; 9: 953211, 2022.
Article in English | MEDLINE | ID: mdl-36299872

ABSTRACT

Background: Mechanisms contributing to tissue remodeling of the infarcted heart following cell-based therapy remain elusive. While cell-based interventions have the potential to influence the cardiac healing process, there is little direct evidence of preservation of functional myocardium. Aim: The aim of the study was to investigate tissue remodeling in the infarcted heart following human embryonic stem cell-derived endothelial cell product (hESC-ECP) therapy. Materials and methods: Following coronary artery ligation (CAL) to induce cardiac ischemia, we investigated infarct size at 1 day post-injection in media-injected controls (CALM, n = 11), hESC-ECP-injected mice (CALC, n = 10), and dead hESC-ECP-injected mice (CALD, n = 6); echocardiography-based functional outcomes 14 days post-injection in experimental (CALM, n = 13; CALC, n = 17) and SHAM surgical mice (n = 4); and mature infarct size (CALM and CALC, both n = 6). We investigated ligand-receptor interactions (LRIs) in hESC-ECP cell populations, incorporating a publicly available C57BL/6J mouse cardiomyocyte-free scRNAseq dataset with naive, 1 day, and 3 days post-CAL hearts. Results: Human embryonic stem cell-derived endothelial cell product injection reduces the infarct area (CALM: 54.5 ± 5.0%, CALC: 21.3 ± 4.9%), and end-diastolic (CALM: 87.8 ± 8.9 uL, CALC: 63.3 ± 2.7 uL) and end-systolic ventricular volume (CALM: 56.4 ± 9.3 uL, CALC: 33.7 ± 2.6 uL). LRI analyses indicate an alternative immunomodulatory effect mediated via viable hESC-ECP-resident signaling. Conclusion: Delivery of the live hESC-ECP following CAL modulates the wound healing response during acute pathological remodeling, reducing infarct area, and preserving functional myocardium in this relatively acute model. Potential intrinsic myocardial cellular/hESC-ECP interactions indicate that discreet immunomodulation could provide novel therapeutic avenues to improve cardiac outcomes following myocardial infarction.

4.
Front Cardiovasc Med ; 8: 795823, 2021.
Article in English | MEDLINE | ID: mdl-35097015

ABSTRACT

Background: Critical limb ischaemia (CLI), which is estimated to affect 2 million people in the United States, reduces quality of life, is associated with high morbidity and mortality, and has limited treatment options. Direct stimulation of angiogenesis using proangiogenic growth factors has been investigated as a therapeutic strategy to improve reperfusion in the ischaemic leg. Despite positive outcomes in animal studies, there has been little success in clinical translation. This investigation addressed the hypothesis that angiogenesis could be stimulated indirectly in the ischaemic hindlimb by blocking 11ß-hydroxysteroid dehydrogenase 1 (11ßHSD1)-mediated reactivation of anti-angiogenic glucocorticoids. Method and Results: Corticosterone suppressed ex vivo angiogenesis in the mouse aortic ring assay. 11ßHSD1 deletion (Hsd11b1Del1/Del1) or pharmacological inhibition (with 300 nM UE2316) which block the reactivation of glucocorticoid (i.e., the conversion of 11-dehydrocorticosterone (11DHC) to bioactive corticosterone) significantly reduced 11DHC-induced suppression of angiogenesis. In a sponge implantation model, 11ßHSD1 deletion, but not pharmacological inhibition, enhanced inflammation-induced angiogenesis. By contrast, in the mouse hindlimb ischaemia model, post-ischaemic reperfusion and vascular density were not affected by either deletion or pharmacological inhibition of 11ßHSD1 in young or aged mice. 3D vascular imaging suggested that hind limb reperfusion in the 1st week following induction of ischaemia may be driven by the rapid expansion of collateral arteries rather than by angiogenesis. Conclusion: 11ßHSD1-mediated glucocorticoid reactivation suppressed angiogenesis ex vivo and in vivo. However, regulation of angiogenesis alone was insufficient to promote reperfusion in hindlimb ischaemia. Future investigation of post-ischaemic reperfusion should include other aspects of systemic vascular remodeling including arteriogenesis and collateral formation.

5.
Physiother Theory Pract ; 33(3): 245-253, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28339334

ABSTRACT

BACKGROUND AND PURPOSE: There are several systems of classification and treatment of patients with low back pain (LBP) based on assessment of the effect of lumbar postures and movements on symptoms. The efficacy of one of these systems, The Movement System Impairment (MSI) method, has not yet been demonstrated in the literature. The purpose of this case report is to describe the approach of the MSI method for an individual with lumbar radiculopathy. CASE DESCRIPTION: A 79-year-old woman with a history of chronic LBP was referred to PT with a physician's diagnosis of sciatica. The patient was classified utilizing a standardized MSI evaluation. She was instructed to modify her daily postures and movements, as well as perform specific exercises to address these impairments. OUTCOMES: Her Oswestry LBP disability score improved by over 30% and pain level per the NPRS improved by 3 out of 10 points. DISCUSSION: Despite the challenges of advanced joint degeneration and neurological involvement, this approach of identifying and addressing specific movement impairments appeared helpful for this older individual.


Subject(s)
Low Back Pain/diagnosis , Low Back Pain/therapy , Lumbar Vertebrae/physiopathology , Motor Activity , Physical Therapy Modalities , Radiculopathy/diagnosis , Radiculopathy/therapy , Sciatica/diagnosis , Sciatica/therapy , Aged , Biomechanical Phenomena , Disability Evaluation , Female , Hip Joint/physiopathology , Humans , Low Back Pain/physiopathology , Pain Measurement , Posture , Predictive Value of Tests , Radiculopathy/physiopathology , Sciatica/physiopathology , Treatment Outcome
6.
Hypertension ; 69(2): 275-285, 2017 02.
Article in English | MEDLINE | ID: mdl-28028193

ABSTRACT

The role of smooth muscle endothelinB (ETB) receptors in regulating vascular function, blood pressure (BP), and neointimal remodeling has not been established. Selective knockout mice were generated to address the hypothesis that loss of smooth muscle ETB receptors would reduce BP, alter vascular contractility, and inhibit neointimal remodeling. ETB receptors were selectively deleted from smooth muscle by crossing floxed ETB mice with those expressing cre-recombinase controlled by the transgelin promoter. Functional consequences of ETB deletion were assessed using myography. BP was measured by telemetry, and neointimal lesion formation induced by femoral artery injury. Lesion size and composition (day 28) were analyzed using optical projection tomography, histology, and immunohistochemistry. Selective deletion of ETB was confirmed by genotyping, autoradiography, polymerase chain reaction, and immunohistochemistry. ETB-mediated contraction was reduced in trachea, but abolished from mesenteric veins, of knockout mice. Induction of ETB-mediated contraction in mesenteric arteries was also abolished in these mice. Femoral artery function was unaltered, and baseline BP modestly elevated in smooth muscle ETB knockout compared with controls (+4.2±0.2 mm Hg; P<0.0001), but salt-induced and ETB blockade-mediated hypertension were unaltered. Circulating endothelin-1 was not altered in knockout mice. ETB-mediated contraction was not induced in femoral arteries by incubation in culture medium or lesion formation, and lesion size was not altered in smooth muscle ETB knockout mice. In the absence of other pathology, ETB receptors in vascular smooth muscle make a small but significant contribution to ETB-dependent regulation of BP. These ETB receptors have no effect on vascular contraction or neointimal remodeling.


Subject(s)
Blood Pressure/physiology , Gene Expression Regulation , Hypertension/genetics , Muscle, Smooth, Vascular/metabolism , RNA/genetics , Receptor, Endothelin B/genetics , Vasoconstriction/physiology , Animals , Cells, Cultured , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Neointima , Real-Time Polymerase Chain Reaction , Receptor, Endothelin B/biosynthesis , Vascular Remodeling
7.
J Vis Exp ; (99): e50627, 2015 May 26.
Article in English | MEDLINE | ID: mdl-26067588

ABSTRACT

The generation and analysis of vascular lesions in appropriate animal models is a cornerstone of research into cardiovascular disease, generating important information on the pathogenesis of lesion formation and the action of novel therapies. Use of atherosclerosis-prone mice, surgical methods of lesion induction, and dietary modification has dramatically improved understanding of the mechanisms that contribute to disease development and the potential of new treatments. Classically, analysis of lesions is performed ex vivo using 2-dimensional histological techniques. This article describes application of optical projection tomography (OPT) to 3-dimensional quantitation of arterial lesions. As this technique is non-destructive, it can be used as an adjunct to standard histological and immunohistochemical analyses. Neointimal lesions were induced by wire-insertion or ligation of the mouse femoral artery whilst atherosclerotic lesions were generated by administration of an atherogenic diet to apoE-deficient mice. Lesions were examined using OPT imaging of autofluorescent emission followed by complementary histological and immunohistochemical analysis. OPT clearly distinguished lesions from the underlying vascular wall. Lesion size was calculated in 2-dimensional sections using planimetry, enabling calculation of lesion volume and maximal cross-sectional area. Data generated using OPT were consistent with measurements obtained using histology, confirming the accuracy of the technique and its potential as a complement (rather than alternative) to traditional methods of analysis. This work demonstrates the potential of OPT for imaging atherosclerotic and neointimal lesions. It provides a rapid, much needed ex vivo technique for the routine 3-dimensional quantification of vascular remodelling.


Subject(s)
Atherosclerosis/pathology , Disease Models, Animal , Tomography, Optical/methods , Animals , Atherosclerosis/etiology , Femoral Artery/pathology , Femoral Artery/surgery , Imaging, Three-Dimensional/methods , Male , Mice , Mice, Inbred C57BL
8.
Br J Pharmacol ; 172(11): 2827-37, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25598351

ABSTRACT

BACKGROUND AND PURPOSE: Endothelin (ET) receptor antagonism reduces neointimal lesion formation in animal models. This investigation addressed the hypothesis that the selective ETA receptor antagonist sitaxentan would be more effective than mixed ETA / B receptor antagonism at inhibiting neointimal proliferation in a mouse model of intraluminal injury. EXPERIMENTAL APPROACH: Antagonism of ETA receptors by sitaxentan (1-100 nM) was assessed in femoral arteries isolated from adult, male C57Bl6 mice using isometric wire myography. Neointimal lesion development was induced by intraluminal injury in mice receiving sitaxentan (ETA antagonist; 15 mg·kg(-1) ·day(-1) ), A192621 (ETB antagonist; 30 mg·kg(-1) ·day(-1) ), the combination of both antagonists or vehicle. Treatment began 1 week before, and continued for 28 days after, surgery. Femoral arteries were then harvested for analysis of lesion size and composition. KEY RESULTS: Sitaxentan produced a selective, concentration-dependent parallel rightward shift of ET-1-mediated contraction in isolated femoral arteries. Sitaxentan reduced neointimal lesion size, whereas ETB and combined ETA / B receptor antagonism did not. Macrophage and α-smooth muscle actin content were unaltered by ET receptor antagonism but sitaxentan reduced the amount of collagen in lesions. CONCLUSIONS AND IMPLICATIONS: These results suggest that ETA receptor antagonism would be more effective than combined ETA /ETB receptor antagonism at reducing neointimal lesion formation.


Subject(s)
Endothelin A Receptor Antagonists/pharmacology , Endothelin B Receptor Antagonists/pharmacology , Femoral Artery/drug effects , Isoxazoles/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Neointima/pathology , Pyrrolidines/pharmacology , Thiophenes/pharmacology , Animals , Disease Models, Animal , Femoral Artery/injuries , Femoral Artery/pathology , Male , Mice , Mice, Inbred C57BL , Myography
9.
Cardiovasc Res ; 103(2): 281-90, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24903497

ABSTRACT

AIMS: Low androgen levels have been linked with an increased risk of cardiovascular disease in men. Previous studies have suggested that androgens directly inhibit atherosclerotic lesion formation although the underlying mechanisms for this remain unclear. This study addressed the hypothesis that endogenous androgens inhibit arterial remodelling by a direct action on the androgen receptor (AR) in the vascular wall. METHODS AND RESULTS: We studied a series of novel mouse lines with cell-specific deletion of the AR in either the endothelium or in smooth muscle cells or both cell types. Findings were compared with a model of global androgen deficiency in wild-type mice (castrated). We characterized the cardiovascular phenotype, vascular pharmacology and histology, and assessed neointimal lesion formation following vascular injury to the femoral artery. Cell-specific AR deletion did not alter body weight, circulating testosterone levels or seminal vesicle weight, but caused limited alterations in arterial contractility and blood pressure. Neointimal lesion formation was unaltered by selective deletion of AR from the vascular endothelium, smooth muscle, or both cell types. Castration in wild-type mice increased neointimal lesion volume (Sham vs. Castration: 2.4 × 10(7) ± 4.5 × 10(6) vs. 3.9 × 10(7) ± 4.9 × 10(6) µm(3), P = 0.04, n = 9-10). CONCLUSION: Vascular cell-specific AR deletion had no effect on neointimal lesion formation, while low systemic androgen levels adversely affect neointimal lesion size. These findings suggest that the cardio-protective effects of androgens are mediated either by AR outside the vasculature or by AR-independent mechanisms.


Subject(s)
Androgens/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Receptors, Androgen/metabolism , Vascular System Injuries/metabolism , Animals , Arteries/metabolism , Arteries/pathology , Blood Pressure/physiology , Cell Proliferation/drug effects , Disease Models, Animal , Mice, Inbred C57BL
10.
FASEB J ; 27(4): 1519-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303209

ABSTRACT

11ß-Hydroxysteroid dehydrogenase type-1 (11ß-HSD1) converts inert cortisone into active cortisol, amplifying intracellular glucocorticoid action. 11ß-HSD1 deficiency improves cardiovascular risk factors in obesity but exacerbates acute inflammation. To determine the effects of 11ß-HSD1 deficiency on atherosclerosis and its inflammation, atherosclerosis-prone apolipoprotein E-knockout (ApoE-KO) mice were treated with a selective 11ß-HSD1 inhibitor or crossed with 11ß-HSD1-KO mice to generate double knockouts (DKOs) and challenged with an atherogenic Western diet. 11ß-HSD1 inhibition or deficiency attenuated atherosclerosis (74-76%) without deleterious effects on plaque structure. This occurred without affecting plasma lipids or glucose, suggesting independence from classical metabolic risk factors. KO plaques were not more inflamed and indeed had 36% less T-cell infiltration, associated with 38% reduced circulating monocyte chemoattractant protein-1 (MCP-1) and 36% lower lesional vascular cell adhesion molecule-1 (VCAM-1). Bone marrow (BM) cells are key to the atheroprotection, since transplantation of DKO BM to irradiated ApoE-KO mice reduced atherosclerosis by 51%. 11ß-HSD1-null macrophages show 76% enhanced cholesterol ester export. Thus, 11ß-HSD1 deficiency reduces atherosclerosis without exaggerated lesional inflammation independent of metabolic risk factors. Selective 11ß-HSD1 inhibitors promise novel antiatherosclerosis effects over and above their benefits for metabolic risk factors via effects on BM cells, plausibly macrophages.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency , Atherosclerosis/metabolism , Bone Marrow/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Atherosclerosis/genetics , Bone Marrow/drug effects , Glucocorticoids/metabolism , Mice , Mice, Knockout , Risk Factors , Vascular Cell Adhesion Molecule-1/metabolism
11.
Cardiovasc Res ; 95(1): 19-28, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22467315

ABSTRACT

AIMS: The potent vasoconstrictor endothelin-1 (ET-1), acting on the endothelin-A (ETA) receptor, promotes intimal lesion formation following vascular injury. The endothelin-B (ETB) receptor, which mediates nitric oxide release and ET-1 clearance in endothelial cells, may moderate lesion formation, but this is less clear. We used selective ET receptor antagonists and cell-specific deletion to address the hypothesis that ETB receptors in the endothelium inhibit lesion formation following arterial injury. METHODS AND RESULTS: Neointimal proliferation was induced by wire or ligation injury to the femoral artery in mice treated with selective ETA (ABT-627) and/or ETB antagonists (A192621). Measurement of lesion formation by optical projection tomography and histology indicated that ETA blockade reduced lesion burden in both models. Although ETB blockade had little effect on ligation injury-induced lesion formation, after wire injury, blockade of the ETB receptor increased lesion burden (184% of vehicle; P < 0.05) and reversed the protective effects of an ETA antagonist. Selective deletion of ETB receptors from the endothelium, however, had no effect on neointimal lesion size. CONCLUSION: These results are consistent with ETB receptor activation playing an important role in limiting neointimal lesion formation following acute vascular injury, but indicate that this protective effect is not mediated by those ETB receptors expressed by endothelial cells. These data support the proposal that selective ETA antagonists may be preferable to mixed ETA/ETB antagonists for targeting the arterial response to injury.


Subject(s)
Neointima/prevention & control , Receptor, Endothelin B/physiology , Vascular System Injuries/pathology , Animals , Blood Pressure/drug effects , Endothelin B Receptor Antagonists , Endothelin-1/pharmacology , Male , Mice , Mice, Inbred C57BL , Pyrrolidines/pharmacology , Receptor, Endothelin A/physiology
12.
J Biol Chem ; 287(6): 4188-97, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22158867

ABSTRACT

In obesity, rapidly expanding adipose tissue becomes hypoxic, precipitating inflammation, fibrosis, and insulin resistance. Compensatory angiogenesis may prevent these events. Mice lacking the intracellular glucocorticoid-amplifying enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ßHSD1(-/-)) have "healthier" adipose tissue distribution and resist metabolic disease with diet-induced obesity. Here we show that adipose tissues of 11ßHSD1(-/-) mice exhibit attenuated hypoxia, induction of hypoxia-inducible factor (HIF-1α) activation of the TGF-ß/Smad3/α-smooth muscle actin (α-SMA) signaling pathway, and fibrogenesis despite similar fat accretion with diet-induced obesity. Moreover, augmented 11ßHSD1(-/-) adipose tissue angiogenesis is associated with enhanced peroxisome proliferator-activated receptor γ (PPARγ)-inducible expression of the potent angiogenic factors VEGF-A, apelin, and angiopoietin-like protein 4. Improved adipose angiogenesis and reduced fibrosis provide a novel mechanism whereby suppression of intracellular glucocorticoid regeneration promotes safer fat expansion with weight gain.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipose Tissue/enzymology , Hypoxia/enzymology , Neovascularization, Physiologic , Obesity/enzymology , Signal Transduction , Actins/genetics , Actins/metabolism , Adipokines , Adipose Tissue/blood supply , Adipose Tissue/pathology , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Angiopoietins/metabolism , Animals , Apelin , Fibrosis/enzymology , Fibrosis/genetics , Fibrosis/physiopathology , Hypoxia/pathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Obesity/pathology , Obesity/physiopathology , PPAR gamma/genetics , PPAR gamma/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Weight Gain/genetics
13.
Cardiovasc Res ; 90(1): 182-90, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21071432

ABSTRACT

AIMS: The vascular type of Ehlers-Danlos syndrome (EDS IV) is an autosomal-dominant disorder characterized by thin translucent skin and extensive bruising. Patients with EDS IV have reduced life expectancy (median 45-50 years) due to spontaneous rupture of arteries (particularly large arteries) or bowel. EDS IV results from mutation of the COL3A1 gene, which encodes the pro-α(1) chains of type III collagen that is secreted into the extracellular matrix, e.g. by smooth muscle cells. A mouse model of EDS IV produced by targeted ablation of Col3a1 has been of limited use as only 10% of homozygous animals survive to adulthood, whereas heterozygous animals do not die from arterial rupture. We report a novel, exploitable model of EDS IV in a spontaneously generated mouse line. METHODS AND RESULTS: Mice were identified by predisposition to sudden, unexpected death from dissection of the thoracic aorta. Aortic dissection inheritance was autosomal-dominant, presented at an early age (median, 6 weeks) with incomplete penetrance, and had a similar sex ratio bias as EDS IV (2:1, male:female). Molecular genetic analysis demonstrated that the causal mutation is a spontaneous 185 kb deletion, including the promoter region and exons 1-39, of the Col3a1 gene. As in EDS IV, aortic dissection was not associated with elevated blood pressure, aneurysm formation, or infection, but may result from aberrant collagen fibrillogenesis within the aortic wall. CONCLUSION: This novel, exploitable mouse line that faithfully models the vascular aspects of human EDS IV provides an important new tool for advancing understanding of EDS IV and of aortic dissection in general.


Subject(s)
Aortic Aneurysm, Thoracic/genetics , Aortic Dissection/genetics , Collagen Type III/genetics , Ehlers-Danlos Syndrome/genetics , Haploinsufficiency , Sequence Deletion , Aortic Dissection/metabolism , Aortic Dissection/pathology , Aortic Dissection/physiopathology , Animals , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/physiopathology , Blood Pressure , Collagen Type III/metabolism , Disease Models, Animal , Ehlers-Danlos Syndrome/metabolism , Ehlers-Danlos Syndrome/pathology , Ehlers-Danlos Syndrome/physiopathology , Exons , Female , Genetic Predisposition to Disease , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Promoter Regions, Genetic
14.
J Gen Intern Med ; 23(3): 323-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18196349

ABSTRACT

BACKGROUND: Understanding the reasons for overweight and obesity is critical to addressing the obesity epidemic. Often the decision to lose weight is based as much on one's self-perception of being overweight as on inherent health benefits. OBJECTIVE: Examine the relationships between self-reported health and demographic factors and measured health risk status and the misperception of actual weight status. DESIGN: Cross-sectional study of factors associated with self-perceived overweight status in participants who self-selected to participate in stroke risk factor screenings. Participants were asked, "Are you overweight?" before their body mass index (BMI) was determined from measured weight and self-reported height. Demographics including, sex, race, education, and location; and health status variables including level of exercise and history of high blood pressure and cholesterol were collected. RESULTS: Mean BMI for the group was 30 kg/m(2). Most women (53.1%) perceived themselves to be overweight, whereas most men (59.6%) perceived themselves not to be overweight. Factors related to misperception of weight status varied by actual BMI category. Among individuals with normal BMI, sedentary individuals had 63% higher odds of misperceiving themselves as overweight. Sedentary individuals with obese BMI were at 55% reduced odds of misperceiving themselves as normal weight. CONCLUSIONS: Active obese and overweight individuals may be more likely to incorrectly perceive themselves as normal weight, and thus misperceive their risk for stroke. Thus, it is not enough to only counsel individuals to be active. Physicians and other health professionals need to counsel their clients to both be active and to attain and maintain a healthy weight.


Subject(s)
Body Mass Index , Obesity/complications , Self Concept , Stroke/epidemiology , Stroke/etiology , Adolescent , Adult , Age Distribution , Attitude to Health , Body Composition , Body Weight , Cohort Studies , Confidence Intervals , Cross-Sectional Studies , Female , Humans , Incidence , Male , Middle Aged , Obesity/diagnosis , Odds Ratio , Probability , Prognosis , Risk Factors , Sex Distribution , Stroke/physiopathology , Survival Rate
15.
Endocrinology ; 148(1): 166-72, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17008390

ABSTRACT

11beta-hydroxysteroid dehydrogenases (11betaHSDs) catalyze interconversion of 11-hydroxy-glucocorticoids with inactive 11-keto metabolites. In blood vessel walls, loss of 11betaHSD1 is thought to reduce local glucocorticoid concentrations, reducing the progression of atheroma and enhancing angiogenesis. Conversely, on the basis that 11betaHSD1 is up-regulated approximately 5-fold by inflammatory cytokines in cultured human vascular smooth muscle cells, it has been proposed that increased 11betaHSD1 during vascular inflammation provides negative feedback suppression of inflammation. We aimed to determine whether inflammation and injury selectively up-regulate 11betaHSD1 reductase activity in vitro and in vivo in intact vascular tissue in mice. In isolated mouse aortae and femoral arteries, reductase activity (converting 11-dehydrocorticosterone to corticosterone) was approximately 10-fold higher than dehydrogenase activity and was entirely accounted for by 11betaHSD1 because it was abolished in vessels from 11betaHSD1(-/-) mice. Although 11betaHSD1 activity was up-regulated by proinflammatory cytokines in cultured murine aortic smooth muscle cells, no such effect was evident in intact aortic rings in vitro. Moreover, after systemic inflammation induced by ip lipopolysaccharide injection, there was only a modest (18%) increase in 11beta-reductase activity in the aorta and no increase in the perfused hindlimb. Furthermore, in femoral arteries in which neointimal proliferation was induced by intraluminal injury, there was no change in basal 11betaHSD1 activity or the sensitivity of 11betaHSD1 to cytokine up-regulation. We conclude that increased generation of glucocorticoids by 11betaHSD1 in the murine vessel wall is unlikely to contribute to feedback regulation of inflammation.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Glucocorticoids/metabolism , Vasculitis/immunology , Vasculitis/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Animals , Aorta/cytology , Aorta/enzymology , Aorta/immunology , Cells, Cultured , Enzyme Activation/drug effects , Femoral Artery/enzymology , Femoral Artery/immunology , Hindlimb/blood supply , Interleukin-1beta/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/immunology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...