Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Nutrients ; 11(11)2019 Nov 02.
Article En | MEDLINE | ID: mdl-31684015

The consumption of fiber-rich foods may negate the deleterious effects of high-fat meals on postprandial triglyceridemia and endothelial function. Despite supportive data in adults, little is known about the effects of high-fat and high-fiber foods on cardiovascular health parameters in pediatric populations. In this crossover trial, male and female adolescents (n = 10; 14.1 + 2.6 years; range 10-17 years) consumed (1) low-fat, low-fiber, (2) low-fat, high-fiber, (3) high-fat, low-fiber, and (4) high-fat, high-fiber breakfast meals in randomized order, each following an overnight fast. Baseline and 4 h post-meal blood was obtained for determination of glucose, insulin and triglyceride concentrations. Endothelial function was assessed via brachial artery flow-mediated dilation (FMD). Postprandial FMD was not significantly changed after any meal. However, regression analyses revealed a significant inverse relationship between the change in 4 h triglyceride concentration and change in 4 h FMD for the high-fat, low-fiber meal (ß = -0.087; 95% CI = -0.138 to -0.037; p = 0.001) that was no longer significant in the high-fat, high-fiber meal (ß = -0.044; 95% CI = -0.117 to 0.029; p = 0.227). Interpretation of these analyses must be qualified by acknowledging that between-meal comparison revealed that the two regression lines were not statistically different (p = 0.226). Addition of high-fiber cereal to the high-fat meal also reduced 4 h postprandial triglyceride increases by ~50% (p = 0.056). A high-fiber breakfast cereal did not attenuate postprandial glucose and insulin responses after consumption of a low-fat meal. While further work is needed to confirm these results in larger cohorts, our findings indicate the potential importance of cereal fiber in blunting the inverse relationship between postprandial hypertriglyceridemia and FMD after consumption of a high-fat meal in adolescents.


Blood Glucose , Dietary Fats/metabolism , Dietary Fiber/pharmacology , Endothelium, Vascular , Triglycerides/blood , Adolescent , Blood Glucose/analysis , Blood Glucose/drug effects , Child , Cross-Over Studies , Diet, Fat-Restricted , Diet, High-Fat , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Male , Postprandial Period/drug effects , Postprandial Period/physiology
2.
S D Med ; 70(7): 319-321, 2017 Jul.
Article En | MEDLINE | ID: mdl-28806029

Carbon monoxide (CO) intoxication is one of the major public health hazards which may go unnoticed as this is a colorless, odorless and tasteless gas. The manifestations of the CO poisoning are far-reaching. Although CO affects almost every organ in the body, cerebral and myocardial involvement are predominant due to the hypoxia-induced cellular damage. The mainstay treatment is providing high-flow oxygen and in some instances hyperbaric oxygen therapy. In the literature, there have been few cases of CO poisoning-induced atrial fibrillation (AF) reported. We hereby report an AF caused by CO toxicity in a young male patient and successful conversion to sinus rhythm with the hyperbaric therapy.


Atrial Fibrillation/etiology , Atrial Fibrillation/therapy , Carbon Monoxide Poisoning/complications , Carbon Monoxide Poisoning/therapy , Hyperbaric Oxygenation , Humans , Hypoxia/complications , Male , Public Health , Young Adult
3.
Am J Physiol Heart Circ Physiol ; 309(6): H1059-65, 2015 Sep 15.
Article En | MEDLINE | ID: mdl-26254335

Animal studies suggest that hypertension leads to cardiac tissue hypothyroidism, a condition that can by itself lead to heart failure. We have previously shown that short-term thyroid hormone treatment in Spontaneously Hypertensive Heart Failure (SHHF) rats near heart failure is beneficial. This study tested the hypothesis that therapeutic, long-term T3 treatment in SHHF rats can prevent or attenuate cardiac dysfunction. Female SHHF rats were treated orally with a physiological T3 dose (0.04 µg/ml) from 12 to 24 mo of age. Age-matched female SHHF and Wistar-Kyoto rats served as hypertensive and normotensive controls, respectively. SHHF rats had reduced serum free thyroid hormone levels and cardiac tissue T3 levels, LV dysfunction, and elevated LV collagen content compared with normotensive controls. Restoration of serum and cardiac tissue thyroid hormone levels in T3-treated rats was associated with no change in heart rate, but strong trends for improvement in LV systolic function and collagen levels. For instance, end-systolic diameter, fractional shortening, systolic wall stress, and LV collagen levels were no longer significantly different from controls. In conclusion, longstanding hypertension in rats led to chronic low serum and cardiac tissue thyroid hormone levels. Long-term treatment with low-dose T3 was safe. While cardiac dysfunction could not be completely prevented in the absence of antihypertensive treatment, T3 may offer additional benefits as an adjunct therapy with possible improvement in diastolic function.


Collagen/drug effects , Heart Failure/etiology , Heart Ventricles/drug effects , Heart/drug effects , Hypertension/complications , Hypothyroidism/etiology , Triiodothyronine/pharmacology , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/drug effects , Animals , Collagen/metabolism , Female , Heart Failure/metabolism , Heart Ventricles/metabolism , Hypertension/metabolism , Hypothyroidism/metabolism , Myosins/drug effects , Myosins/metabolism , Random Allocation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Thyroxine/metabolism , Ventricular Dysfunction, Left/metabolism
4.
Mol Med ; 20: 302-12, 2014 May 01.
Article En | MEDLINE | ID: mdl-24960246

Thyroid dysfunction is common in individuals with diabetes mellitus (DM) and may contribute to the associated cardiac dysfunction. However, little is known about the extent and pathophysiological consequences of low thyroid conditions on the heart in DM. DM was induced in adult female Sprague Dawley (SD) rats by injection of nicotinamide (N; 200 mg/kg) followed by streptozotocin (STZ; 65 mg/kg). One month after STZ/N, rats were randomized to the following groups (N = 10/group): STZ/N or STZ/N + 0.03 µg/mL T3; age-matched vehicle-treated rats served as nondiabetic controls (C). After 2 months of T3 treatment (3 months post-DM induction), left ventricular (LV) function was assessed by echocardiography and LV pressure measurements. Despite normal serum thyroid hormone (TH) levels, STZ/N treatment resulted in reductions in myocardial tissue content of THs (T3 and T4: 39% and 17% reduction versus C, respectively). Tissue hypothyroidism in the DM hearts was associated with increased DIO3 deiodinase (which converts THs to inactive metabolites) altered TH transporter expression, reexpression of the fetal gene phenotype, reduced arteriolar resistance vessel density, and diminished cardiac function. Low-dose T3 replacement largely restored cardiac tissue TH levels (T3 and T4: 43% and 10% increase versus STZ/N, respectively), improved cardiac function, reversed fetal gene expression and preserved the arteriolar resistance vessel network without causing overt symptoms of hyperthyroidism. We conclude that cardiac dysfunction in chronic DM may be associated with tissue hypothyroidism despite normal serum TH levels. Low-dose T3 replacement appears to be a safe and effective adjunct therapy to attenuate and/or reverse cardiac remodeling and dysfunction induced by experimental DM.


Diabetes Mellitus, Experimental/drug therapy , Hormone Replacement Therapy , Myocardium/metabolism , Thyroid Hormones/therapeutic use , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Hemodynamics , Myocardium/pathology , Rats, Sprague-Dawley , Thyroid Hormones/blood , Thyroid Hormones/pharmacology , Ventricular Remodeling/drug effects
5.
Circ Arrhythm Electrophysiol ; 6(5): 952-9, 2013 Oct.
Article En | MEDLINE | ID: mdl-24036190

BACKGROUND: Evidence indicates that cardiac hypothyroidism may contribute to heart failure progression. It is also known that heart failure is associated with an increased risk of atrial fibrillation (AF). Although it is established that hyperthyroidism increases AF incidence, the effect of hypothyroidism on AF is unclear. This study investigated the effects of different thyroid hormone levels, ranging from hypothyroidism to hyperthyroidism on AF inducibility in thyroidectomized rats. METHODS AND RESULTS: Thyroidectomized rats with serum-confirmed hypothyroidism 1 month after surgery were randomized into hypothyroid (N=9), euthyroid (N=9), and hyperthyroid (N=9) groups. Rats received placebo, 3.3-mg l-thyroxine (T4), or 20-mg T4 pellets (60-day release form) for 2 months, respectively. At the end of treatment, hypothyroid, euthyroid, and hyperthyroid status was confirmed. Hypothyroid animals showed cardiac atrophy and reduced cardiac systolic and diastolic functions, whereas hyperthyroid rats exhibited cardiac hypertrophy and increased cardiac function. Hypothyroidism and hyperthyroidism produced opposite electrophysiological changes in heart rates and atrial effective refractory period, but both significantly increased AF susceptibility. AF incidence was 78% in hypothyroid, 67% in hyperthyroid, and the duration of induced AF was also longer, compared with 11% in the euthyroid group (all P<0.05). Hypothyroidism increased atrial interstitial fibrosis, but connexin 43 was not affected. CONCLUSIONS: Both hypothyroidism and hyperthyroidism lead to increased AF vulnerability in a rat thyroidectomy model. Our results stress that normal thyroid hormone levels are required to maintain normal cardiac electrophysiology and to prevent cardiac arrhythmias and AF.


Atrial Fibrillation/etiology , Hyperthyroidism/complications , Hypothyroidism/complications , Animals , Atrial Fibrillation/diagnostic imaging , Atrial Fibrillation/physiopathology , Disease Models, Animal , Echocardiography , Electrophysiologic Techniques, Cardiac , Enzyme-Linked Immunosorbent Assay , Female , Hemodynamics , Immunohistochemistry , Random Allocation , Rats , Rats, Sprague-Dawley , Thyroid Hormones/blood , Thyroidectomy
7.
Endocrinology ; 154(7): 2542-52, 2013 Jul.
Article En | MEDLINE | ID: mdl-23594789

Thyroid hormones (THs) play a pivotal role in regulating cardiovascular homeostasis. To provide a better understanding of the coordinated processes that govern cardiac TH bioavailability, this study investigated the influence of serum and cardiac TH status on the expression of TH transporters and cytosolic binding proteins in the myocardium. In addition, we sought to determine whether the administration of T(3) (instead of T(4)) improves the relationship between THs in serum and cardiac tissue and cardiac function over a short-term treatment period. Adult female Sprague Dawley rats were made hypothyroid by 7 weeks treatment with the antithyroid drug 6-n-propyl-2-thiouracil (PTU). After establishing hypothyroidism, rats were assigned to 1 of 5 graded T(3) dosages plus PTU for a 2-week dose-response experiment. Untreated, age-matched rats served as euthyroid controls. PTU was associated with depressed serum and cardiac tissue T(3) and T(4) levels, arteriolar atrophy, altered TH transporter and cytosolic TH binding protein expression, fetal gene reexpression, and cardiac dysfunction. Short-term administration of T(3) led to a mismatch between serum and cardiac tissue TH levels. Normalization of serum T(3) levels was not associated with restoration of cardiac tissue T(3) levels or cardiac function. In fact, a 3-fold higher T(3) dosage was necessary to normalize cardiac tissue T(3) levels and cardiac function. Importantly, this study provides the first comprehensive data on the relationship between altered TH status (serum and cardiac tissue), cardiac function, and the coordinated in vivo changes in cardiac TH membrane transporters and cytosolic TH binding proteins in altered TH states.


Hypothyroidism/drug therapy , Thyroid Hormones/therapeutic use , Animals , Disease Models, Animal , Female , Heart/drug effects , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Thyroid Hormones/blood , Thyroxine/blood , Thyroxine/therapeutic use , Triiodothyronine/blood , Triiodothyronine/therapeutic use
8.
J Transl Med ; 11: 40, 2013 Feb 14.
Article En | MEDLINE | ID: mdl-23409791

BACKGROUND: Left ventricular (LV) remodeling following large transmural myocardial infarction (MI) remains a pivotal clinical issue despite the advance of medical treatment over the past few decades. Identification of new medications to improve the remodeling process and prevent progression to heart failure after MI is critical. Thyroid hormones (THs) have been shown to improve LV function and remodeling in animals post-MI and in the human setting. However, changes in underlying cellular remodeling resulting from TH treatment are not clear. METHODS: MI was produced in adult female Sprague-Dawley rats by ligation of the left descending coronary artery. L-thyroxine (T4) pellet (3.3 mg, 60 days sustained release) was used to treat MI rats for 8 weeks. Isolated myocyte shape, arterioles, and collagen deposition in the non-infarcted area were measured at terminal study. RESULTS: T4 treatment improved LV ±dp/dt, normalized TAU, and increased myocyte cross-sectional area without further increasing myocyte length in MI rats. T4 treatment increased the total LV tissue area by 34%, increased the non-infarcted tissue area by 41%, and increased the thickness of non-infarcted area by 36% in MI rats. However, myocyte volume accounted for only ~1/3 of the increase in myocyte mass in the non-infarct area, indicating the presence of more myocytes with treatment. T4 treatment tended to increase the total length of smaller arterioles (5 to 15 µm) proportional to LV weight increase and also decreased collagen deposition in the LV non-infarcted area. A tendency for increased metalloproteinase-2 (MMP-2) expression and tissue inhibitor of metalloproteinases (TIMPs) -1 to -4 expression was also observed in T4 treated MI rats. CONCLUSIONS: These results suggest that long-term T4 treatment after MI has beneficial effects on myocyte, arteriolar, and collagen matrix remodeling in the non-infarcted area. Most importantly, results suggest improved survival of myocytes in the peri-infarct area.


Myocardial Infarction/pathology , Thyroxine/administration & dosage , Ventricular Remodeling/drug effects , Animals , Arterioles/pathology , Body Weight/drug effects , Collagen/metabolism , Echocardiography , Female , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Thyroxine/blood , Triiodothyronine/blood
9.
PLoS One ; 7(10): e46655, 2012.
Article En | MEDLINE | ID: mdl-23056390

Thyroid hormones (THs) play a pivotal role in cardiac homeostasis. TH imbalances alter cardiac performance and ultimately cause cardiac dysfunction. Although short-term hyperthyroidism typically leads to heightened left ventricular (LV) contractility and improved hemodynamic parameters, chronic hyperthyroidism is associated with deleterious cardiac consequences including increased risk of arrhythmia, impaired cardiac reserve and exercise capacity, myocardial remodeling, and occasionally heart failure. To evaluate the long-term consequences of chronic hyperthyroidism on LV remodeling and function, we examined LV isolated myocyte function, chamber function, and whole tissue remodeling in a hamster model. Three-month-old F1b hamsters were randomized to control or 10 months TH treatment (0.1% grade I desiccated TH). LV chamber remodeling and function was assessed by echocardiography at 1, 2, 4, 6, 8, and 10 months of treatment. After 10 months, terminal cardiac function was assessed by echocardiography and LV hemodynamics. Hyperthyroid hamsters exhibited significant cardiac hypertrophy and deleterious cardiac remodeling characterized by myocyte lengthening, chamber dilatation, decreased relative wall thickness, increased wall stress, and increased LV interstitial fibrotic deposition. Importantly, hyperthyroid hamsters demonstrated significant LV systolic and diastolic dysfunction. Despite the aforementioned remodeling and global cardiac decline, individual isolated cardiac myocytes from chronically hyperthyroid hamsters had enhanced function when compared with myocytes from untreated age-matched controls. Thus, it appears that long-term hyperthyroidism may impair global LV function, at least in part by increasing interstitial ventricular fibrosis, in spite of normal or enhanced intrinsic cardiomyocyte function.


Hyperthyroidism/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cells, Cultured , Cricetinae , Echocardiography , Hyperthyroidism/chemically induced , Myocytes, Cardiac/drug effects , Thyroid Hormones/pharmacology , Ventricular Remodeling/drug effects
10.
PLoS One ; 7(8): e40161, 2012.
Article En | MEDLINE | ID: mdl-22870193

INTRODUCTION: The expression of hundreds of genes is altered in response to left ventricular (LV) remodeling following large transmural myocardial infarction (MI). Thyroid hormone (TH) improves LV remodeling and cardiac performance after MI. However, the molecular basis is unknown. METHODS: MI was produced by ligation of the left anterior descending coronary artery in female SD rats. Rats were divided into the following groups: (1) Sham MI, (2) MI, and (3) MI+T4 treatment (T4 pellet 3.3 mg, 60 days release, implanted subcutaneously immediately following MI). Four weeks after surgery, total RNA was isolated from LV non-infarcted areas for microarray analysis using the Illumina RatRef-12 Expression BeadChip Platform. RESULTS: Signals were detected in 13,188 genes (out of 22,523), of which the expression of 154 genes were decreased and the expression of 200 genes were increased in MI rats compared with Sham MI rats (false discovery rate (FDR) <0.05). Compared to MI rats, T4 treatment decreased expression of 27 genes and increased expression of 28 genes. In particular, 6 genes down-regulated by MI and 12 genes up-regulated by MI were reversed by T4. Most of the 55 genes altered by T4 treatment are in the category of molecular function under binding (24) and biological processes which includes immune system process (9), multi-organism process (5) and biological regulation (19) nonexclusively. CONCLUSIONS: These results suggest that altered expression of genes for molecular function and biological process may be involved in the beneficial effects of thyroid hormone treatment following MI in rats.


Gene Expression Regulation , Muscle Proteins/biosynthesis , Myocardial Infarction/metabolism , Thyroid Hormones/metabolism , Ventricular Remodeling , Animals , Female , Gene Expression Profiling/methods , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley
11.
PLoS One ; 6(9): e25054, 2011.
Article En | MEDLINE | ID: mdl-21966411

Low thyroid hormone (TH) function has been linked to impaired coronary blood flow, reduced density of small arterioles, and heart failure. Nonetheless, little is known about the mechanisms by which THs regulate coronary microvascular remodeling. The current study examined the initial cellular events associated with coronary remodeling induced by triiodothyronine (T3) in hypothyroid rats. Rats with established hypothyroidism, eight weeks after surgical thyroidectomy (TX), were treated with T3 for 36 or 72 hours. The early effects of T3 treatment on coronary microvasculature were examined morphometrically. Gene expression changes in the heart were assessed by quantitative PCR Array. Hypothyroidism resulted in arteriolar atrophy in the left ventricle. T3 treatment rapidly induced small arteriolar muscularization and, within 72 hours, restored arteriolar density to control levels. Total length of the capillary network was not affected by TX or T3 treatment. T3 treatment resulted in the coordinate regulation of Angiopoietin 1 and 2 expression. The response of Angiopoietins was consistent with vessel enlargement. In addition to the well known effects of THs on vasoreactivity, these results suggest that THs may affect function of small resistance arteries by phenotypic remodeling of vascular smooth muscle cells (VSMC).


Coronary Vessels/physiopathology , Thyroid Hormones/metabolism , Triiodothyronine/metabolism , Angiopoietins/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Echocardiography/methods , Gene Expression Regulation , Heart Failure , Hypothyroidism/metabolism , Microscopy, Fluorescence/methods , Muscle, Smooth, Vascular/cytology , Myocardium/metabolism , Phenotype , Rats , Thyroidectomy/methods , Time Factors , Vascular Resistance
12.
Eur J Appl Physiol ; 111(9): 2063-8, 2011 Sep.
Article En | MEDLINE | ID: mdl-21259025

The purpose of the study was to examine the relationship between VO(2) and RPE at the lactate threshold (LT) and maximal fat oxidation rate (FAT(MAX)) in untrained adults and determine the stability of the relationship across sex, age, and fitness status. A total of 148 untrained adults (mean age [year] = 30.5 ± 13.9, height [m] = 1.72 ± 0.08 m, body mass [kg] = 82.6 ± 20.5, body fat [%] = 28.7 ± 12.0) completed a continuous incremental VO(2) peak/LT protocol. Fat oxidation rates were determined using indirect calorimetry. The highest recorded fat oxidation rate was chosen as FAT(MAX). The breakpoint in the VO(2)-blood lactate relationship was chosen as LT. RPE was based on the Borg 6-20 scale. Bland-Altman plot analysis demonstrated that VO(2) FAT(MAX) systematically preceded VO(2) LT (mean bias = 1.3 ml kg(-1) min(-1)) with wide limits of agreement (+9.6 to -6.9 ml kg(-1) min(-1)). Multivariate ANOVA revealed a significant difference between VO(2) FAT(MAX) (12.7 ± 7.5 ml kg(-1) min(-1)) and VO(2) LT (14.1 ± 5.9 ml kg(-1) min(-1)) in the total sample (p = 0.04). There were no differences between the intensities when the sample was divided into sex, age, and fitness comparison groups (p values >0.05). RPE FAT(MAX) (9.4 ± 2.5) preceded RPE LT (10.4 ± 2.0) in the total sample (p = 0.008), but was not different across comparison groups (p > 0.05). The present data indicate that the highest rate of fat oxidation slightly precedes the LT in untrained adults. For exercise prescription, a Borg-RPE of 9-12 identifies both FAT(MAX) and LT.


Lactic Acid/blood , Lipid Metabolism , Oxygen Consumption/physiology , Perception/physiology , Physical Exertion/physiology , Adolescent , Adult , Fats/metabolism , Female , Humans , Lactic Acid/metabolism , Lipid Metabolism/physiology , Male , Middle Aged , Oxidation-Reduction , Physical Education and Training , Research Design , Sedentary Behavior , Young Adult
13.
Clin Sports Med ; 28(3): 423-39, 2009 Jul.
Article En | MEDLINE | ID: mdl-19505624

Management of diabetes requires a multidisciplinary approach including: medical therapy, nutritional therapy, self-management education, psychosocial assessment and care, hypoglycemia awareness training, and exercise. Exercise in an effective lifestyle management technique for the prevention of type 2 diabetes and for the management of both type 1 diabetes and type 2 diabetes. Here we review the use of exercise evaluation and prescription for the prevention and management of diabetes.


Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/therapy , Exercise/physiology , Adolescent , Adult , Aged , Child , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 2/diagnosis , Exercise Test , Female , Humans , Male , Middle Aged , Young Adult
...