Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1266444, 2023.
Article in English | MEDLINE | ID: mdl-37942229

ABSTRACT

Developmental programming of chronic adverse cardiovascular health outcomes has been studied both using numerous human populations and an array of animal models. However, the mechanisms that produce transgenerational effects have been difficult to study due to a lack of developmentally relevant models. As such, how increased disease risk is carried to the second generation has been poorly studied. We hypothesized that the endothelium which mediates many acute and chronic vascular inflammatory responses is a key player in these effects, and epidemiological studies implicate transgenerational nutritional effects on endothelial health. To study the mutigenerational effects of maternal undernutrition on offspring endothelial health, we developed a model of transgenerational nutritional stress in guinea pigs, a translationally relevant precocial species with a relatively short lifespan. First- and second-generation offspring were subjected to a high fat diet in adolescence to exacerbate negative cardiovascular health. To assess transcriptional changes, we performed bulk RNA-sequencing in carotid artery endothelial cells, with groups stratified as prenatal control or food restricted, and postnatal control or high fat diet. We detected statistically significant gene alterations for each dietary permutation, some of which were unique to treatments and other transcriptional signatures shared by multiple or all conditions. These findings highlight a core group of genes altered by high fat diet that is shared by all cohorts and a divergence of transgenerational effects between the prenatal ad libitum and dietary restriction groups. This study establishes the groundwork for this model to be used to better understand the interplay of prenatal stress and genetic reprogramming.

2.
J Physiol ; 2023 May 02.
Article in English | MEDLINE | ID: mdl-37128962

ABSTRACT

Contraction of cardiomyocytes is initiated at subcellular elements called dyads, where L-type Ca2+ channels in t-tubules are located within close proximity to ryanodine receptors in the sarcoplasmic reticulum. While evidence from small rodents indicates that dyads are assembled gradually in the developing heart, it is unclear how this process occurs in large mammals. We presently examined dyadic formation in fetal and newborn sheep (Ovis aries), and the regulation of this process by fetal cardiac workload. By employing advanced imaging methods, we demonstrated that t-tubule growth and dyadic assembly proceed gradually during fetal sheep development, from 93 days of gestational age until birth (147 days). This process parallels progressive increases in fetal systolic blood pressure, and includes step-wise colocalization of L-type Ca2+ channels and the Na+ /Ca2+ exchanger with ryanodine receptors. These proteins are upregulated together with the dyadic anchor junctophilin-2 during development, alongside changes in the expression of amphiphysin-2 (BIN1) and its partner proteins myotubularin and dynamin-2. Increasing fetal systolic load by infusing plasma or occluding the post-ductal aorta accelerated t-tubule growth. Conversely, reducing fetal systolic load with infusion of enalaprilat, an angiotensin converting enzyme inhibitor, blunted t-tubule formation. Interestingly, altered t-tubule densities did not relate to changes in dyadic junctions, or marked changes in the expression of dyadic regulatory proteins, indicating that distinct signals are responsible for maturation of the sarcoplasmic reticulum. In conclusion, augmenting blood pressure and workload during normal fetal development critically promotes t-tubule growth, while additional signals contribute to dyadic assembly. KEY POINTS: T-tubule growth and dyadic assembly proceed gradually in cardiomyocytes during fetal sheep development, from 93 days of gestational age until the post-natal stage. Increasing fetal systolic load by infusing plasma or occluding the post-ductal aorta accelerated t-tubule growth and hypertrophy. In contrast, reducing fetal systolic load by enalaprilat infusion slowed t-tubule development and decreased cardiomyocyte size. Load-dependent modulation of t-tubule maturation was linked to altered expression patterns of the t-tubule regulatory proteins junctophilin-2 and amphiphysin-2 (BIN1) and its protein partners. Altered t-tubule densities did not influence dyadic formation, indicating that distinct signals are responsible for maturation of the sarcoplasmic reticulum.

3.
Artif Organs ; 45(2): 135-142, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32857895

ABSTRACT

This article describes the properties and performance of a rotary total artificial heart (TAH) that produces inherently pulsatile flow. The hydraulic performance of the TAH was characterized using a mock circulatory loop to simulate four physiologically relevant conditions: baseline flow, increased flow, systemic hypertension, and pulmonary hypertension. The pump has a variable shuttle rate (beats per minute), percentage dwell time, and angular velocity on either side (revolutions per minute), which allows for full control of the flow rate and pulsatility over a range of healthy and pathologic pressures and flow rates. The end-to-end length and displacement volume of the TAH are 9.8 cm and 130 mL, respectively, allowing it to fit in smaller chest cavities including those of smaller adults and juvenile humans.


Subject(s)
Heart Failure/surgery , Heart Ventricles/physiopathology , Heart, Artificial , Models, Cardiovascular , Prosthesis Design , Heart Failure/physiopathology , Humans , Hypertension/physiopathology , Hypertension, Pulmonary/physiopathology , Pulsatile Flow/physiology
4.
FASEB J ; 34(8): 10041-10055, 2020 08.
Article in English | MEDLINE | ID: mdl-32573852

ABSTRACT

As loss of contractile function in heart disease could often be mitigated by increased cardiomyocyte number, expansion of cardiomyocyte endowment paired with increased vascular supply is a desirable therapeutic goal. Insulin-like growth factor 1 (IGF-1) administration increases fetal cardiomyocyte proliferation and heart mass, but how fetal IGF-1 treatment affects coronary growth and function is unknown. Near-term fetal sheep underwent surgical instrumentation and were studied from 127 to 134 d gestation (term = 147 d), receiving either IGF-1 LR3 or vehicle. Coronary growth and function were interrogated using pressure-flow relationships, an episode of acute hypoxia with progressive blockade of adenosine receptors and nitric oxide synthase, and by modeling the determinants of coronary flow. The main findings were that coronary conductance was preserved on a per-gram basis following IGF-1 treatment, adenosine and nitric oxide contributed to hypoxia-mediated coronary vasodilation similarly in IGF-1-treated and Control fetuses, and the relationships between coronary flow and blood oxygen contents were similar between groups. We conclude that IGF-1-stimulated fetal myocardial growth is accompanied by appropriate expansion and function of the coronary vasculature. These findings support IGF-1 as a potential strategy to increase cardiac myocyte and coronary vascular endowment at birth.


Subject(s)
Coronary Vessels/growth & development , Fetus/physiology , Insulin-Like Growth Factor I/pharmacology , Myocytes, Cardiac/physiology , Animals , Coronary Vessels/cytology , Coronary Vessels/drug effects , Female , Fetus/drug effects , Hypoxia/physiopathology , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Sheep
5.
J Physiol ; 597(8): 2163-2176, 2019 04.
Article in English | MEDLINE | ID: mdl-30770568

ABSTRACT

KEY POINTS: Plasma thyroid hormone (tri-iodo-l-thyronine; T3 ) concentrations rise near the end of gestation and is known to inhibit proliferation and stimulate maturation of cardiomyocytes before birth. Thyroid hormone receptors are required for the action of thyroid hormone in fetal cardiomyocytes. Loss of thyroid hormone receptor (TR)α1 abolishes T3 signalling via extracellular signal-related kinase and Akt in fetal cardiomyocytes. The expression of TRα1 and TRß1 in ovine fetal myocardium increases with age, although TRα1 levels always remain higher than those of TRß1. Near term fetal cardiac myocytes are more sensitive than younger myocytes to thyroid receptor blockade by antagonist, NH3, and to the effects of TRα1/α2 short interfering RNA. Although T3 is known to abrogate ovine cardiomyocyte proliferation stimulated by insulin-like growth factor 1, this effect is mediated via the genomic action of thyroid hormone receptors, with little evidence for non-genomic mechanisms. ABSTRACT: We have previously shown that the late-term rise in tri-iodo-l-thyronine (T3 ) in fetal sheep leads to the inhibition of proliferation and promotion of maturation in cardiomyocytes. The present study was designed to determine whether these T3 -induced changes are mediated via thyroid hormone receptors (TRs) or by non-genomic mechanisms. Fetal cardiomyocytes were isolated from 102 ± 3 and 135 ± 1 days of gestational age (dGA) sheep (n = 7 per age; term ∼145 dGA). Cells were treated with T3 (1.5 nm), insulin-like growth factor (IGF)-1 (1 µg mL-1 ) or a combination in the presence of TR antagonist NH3 (100 nm) or following short interfering RNA (siRNA) knockdown of TRα1/α2. Proliferation was quantified by 5-bromo-2'-deoxyuridine (BrdU) uptake (10 µm). Western blots measured protein levels of extracellular signal-related kinase (ERK), Akt, TRα1/ß1 and p21. Age specific levels of TRα1/ß1 were measured in normal hearts from fetuses [95 dGA (n = 8), 135 dGA (n = 7)], neonates (n = 8) and adult ewes (n = 7). TRα1 protein levels were consistently >50% more than TRß1 at each gestational age (P < 0.05). T3 reduced IGF-1 stimulated proliferation by ∼50% in 100 dGA and by ∼75% in 135 dGA cardiomyocytes (P < 0.05). NH3 blocked the T3  + IGF-1 reduction of BrdU uptake without altering the phosphorylation of ERK or Akt at both ages. NH3 did not suppress T3 -induced p21 expression in 100 dGA cardiomyocytes in 135 dGA cardiomyocytes, NH3 alone reduced BrdU uptake (-28%, P < 0.05), as well as T3 -induced p21 (-75%, P < 0.05). In both ages, siRNA knockdown of TRα1/α2 blocked the T3  + IGF-1 reduction of BrdU uptake and dramatically reduced ERK and Akt signalling in 135 dGA cardiomyocytes. In conclusion, TRs are required for normal proliferation and T3 signalling in fetal ovine cardiomyocytes, with the sensitivity to TR blockade being age-dependent.


Subject(s)
Myocytes, Cardiac/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Cell Proliferation , Cells, Cultured , Fetal Heart/cytology , Fetal Heart/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sheep , Triiodothyronine/metabolism
6.
Am J Physiol Heart Circ Physiol ; 316(5): H985-H991, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30707615

ABSTRACT

The fetal myocardium is known to be sensitive to hemodynamic load, responding to systolic overload with cellular hypertrophy, proliferation, and accelerated maturation. However, the fetal cardiac growth response to primary volume overload is unknown. We hypothesized that increased venous return would stimulate fetal cardiomyocyte proliferation and terminal differentiation, particularly in the right ventricle (RV). Vascular catheters and pulmonary artery flow probes were implanted in 16 late-gestation fetal sheep: a right carotid artery-jugular vein (AV) fistula was surgically created in nine fetuses, and sham operations were performed on seven fetuses. Instrumented fetuses were studied for 1 wk before hearts were dissected for component analysis or cardiomyocyte dispersion for cellular measurements. Within 1 day of AV fistula creation, RV output was 20% higher in experimental than sham fetuses ( P < 0.0001). Circulating atrial natriuretic peptide levels were elevated fivefold in fetuses with an AV fistula ( P < 0.002). On the terminal day, RV-to-body weight ratios were 35% higher in the AV fistula group ( P < 0.05). Both left ventricular and RV cardiomyocytes grew longer in fetuses with an AV fistula ( P < 0.02). Cell cycle activity was depressed by >50% [significant in left ventricle ( P < 0.02), but not RV ( P < 0.054)]. Rates of terminal differentiation were unchanged. Based on these studies, we speculate that atrial natriuretic peptide suppressed fetal cardiomyocyte cell cycle activity. Unlike systolic overload, fetal diastolic load appears to drive myocyte enlargement, but not cardiomyocyte proliferation or maturation. These changes could predispose to RV dysfunction later in life. NEW & NOTEWORTHY Adaptation of the fetal heart to changes in cardiac load allows the fetus to maintain adequate blood flow to its systemic and placental circulations, which is necessary for the well-being of the fetus. Addition of arterial-venous fistula flow to existing venous return increased right ventricular stroke volume and output. The fetal heart compensated by cardiomyocyte elongation without accelerated cellular maturation, while cardiomyocyte proliferation decreased. Even transient volume overload in utero alters myocardial structure and cardiomyocyte endowment.


Subject(s)
Fetal Heart/physiopathology , Hypertrophy, Right Ventricular/physiopathology , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right , Ventricular Remodeling , Animals , Arteriovenous Shunt, Surgical , Atrial Natriuretic Factor/blood , Carotid Arteries/surgery , Cell Cycle Checkpoints , Cell Differentiation , Cell Proliferation , Cell Size , Disease Models, Animal , Female , Fetal Heart/metabolism , Fetal Heart/pathology , Gestational Age , Hypertrophy, Right Ventricular/blood , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/pathology , Jugular Veins/surgery , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pregnancy , Sheep, Domestic , Stroke Volume , Ventricular Dysfunction, Right/blood , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/pathology
7.
Am J Physiol Regul Integr Comp Physiol ; 315(5): R1038-R1048, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30480483

ABSTRACT

Although cardiomyocyte terminal differentiation is nearly complete at birth in sheep, as in humans, very limited postnatal expansion of myocyte number may occur. The capacity of newborn cardiomyocytes to respond to growth stimulation by proliferation is poorly understood. Our objective was to test this growth response in newborn lambs with two stimuli shown to be potent inducers of cardiomyocyte growth in fetuses and adults: increased systolic load (Load) and insulin-like growth factor I (IGF-I). Vascular catheters and an inflatable aortic occluder were implanted in lambs. Hearts were collected for analysis at 18 days of age after a 7-day experiment and compared with control hearts. Load hearts, but not IGF-I hearts, were heavier ( P = 0.001) because of increased mass of the left ventricle (LV), septum, and left atrium (40-50%, P = 0.004). Terminal differentiation and cell cycle activity were not different between groups. Myocyte length was 7% greater in Load lamb hearts ( P < 0.05), and binucleated myocytes, which comprise ~90% of LV cells, were 25% larger in volume ( P = 0.03). Myocyte number per gram of myocardium was decreased in all ventricles of Load lambs ( P = 0.01). Cells from the IGF-I group were not different by any comparison. These results suggest that the newborn sheep LV responds to systolic stress with cardiomyocyte hypertrophy, not proliferation. Furthermore, IGF-I is ineffective at stimulating cardiomyocyte proliferation at this age (despite effectiveness when administered before birth). Thus, to expand cardiomyocyte number in the newborn heart, therapies other than systolic pressure load and IGF-I treatment need to be developed.


Subject(s)
Hypertension/complications , Insulin-Like Growth Factor I/pharmacology , Myocytes, Cardiac/drug effects , Recombinant Proteins/drug effects , Animals , Fetus/drug effects , Fetus/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertrophy/drug therapy , Infant, Newborn , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Sheep
8.
Circulation ; 135(7): 700-710, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28193800

ABSTRACT

Although laparoscopic surgery accounts for >2 million surgical procedures every year, the current preoperative risk scores and guidelines do not adequately assess the risks of laparoscopy. In general, laparoscopic procedures have a lower risk of morbidity and mortality compared with operations requiring a midline laparotomy. During laparoscopic surgery, carbon dioxide insufflation may produce significant hemodynamic and ventilatory consequences such as increased intraabdominal pressure and hypercarbia. Hemodynamic insults secondary to increased intraabdominal pressure include increased afterload and preload and decreased cardiac output, whereas ventilatory consequences include increased airway pressures, hypercarbia, and decreased pulmonary compliance. Hemodynamic effects are accentuated in patients with cardiovascular disease such as congestive heart failure, ischemic heart disease, valvular heart disease, pulmonary hypertension, and congenital heart disease. Prevention of cardiovascular complications may be accomplished through a sound understanding of the hemodynamic and physiological consequences of laparoscopic surgery as well as a defined operative plan generated by a multidisciplinary team involving the preoperative consultant, anesthesiologist, and surgeon.


Subject(s)
Blood Pressure/physiology , Heart Rate/physiology , Hemodynamics/physiology , Laparoscopy/adverse effects , Humans , Laparoscopy/methods , Models, Animal
9.
J Physiol ; 594(21): 6165-6174, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27291778

ABSTRACT

KEY POINTS: In fetuses, chronic anaemia stimulates cardiac growth; simultaneously, blood flow to the heart muscle itself is increased, and reserve blood flow capacity of the coronary vascular bed is preserved. Here we examined functional adaptations of the capillaries and small blood vessels responsible for delivering oxygen to the anaemic fetal heart muscle using contrast-enhanced echocardiography. We demonstrate that coronary microvascular flux rate doubled in anaemic fetuses compared to control fetuses, both at rest and during maximal flow, suggesting reduced microvascular resistance consistent with capillary widening. Cardiac fractional microvascular blood volume was not greater in anaemic fetuses, suggesting that growth of new microvascular vessels does not contribute to the increased flow per volume of myocardium. These unusual changes in microvascular function during anaemia may indicate novel adaptive strategies in the fetal heart. ABSTRACT: Fetal anaemia causes cardiac adaptations that have immediate and life-long repercussions on heart function and health. It is known that resting and maximal coronary conductance both increase during chronic fetal anaemia, but the coronary microvascular changes responsible for the adaptive response are unknown. Until recently, technical limitations have prevented quantifying functional capillary-level adaptations in the in vivo fetal heart. Our objective was to characterise functional microvascular adaptations in chronically anaemic fetal sheep. Chronically instrumented fetuses were randomized to a control group (n = 11) or were made anaemic by isovolumetric haemorrhage (n = 12) for 1 week prior to myocardial contrast echocardiography at 85% of gestation. Anaemia augmented cardiac mass by 23% without changing body weight. In anaemic fetuses, microvascular blood flow per volume of myocardium was twice that of control fetuses at rest, during vasodilatory hyperaemia, and during hyperaemia plus increased aortic pressure. The elevated blood flow was attributable almost entirely to an increase in microvascular blood flux rate whereas microvascular blood volumes were not different between groups at baseline, during hyperaemia, or with hyperaemia plus increased aortic pressure. Increased coronary microvascular flux rate in response to chronic fetal anaemia is consistent with expected reductions in capillary resistance from capillary diameter widening detected in earlier histological studies.


Subject(s)
Adaptation, Physiological , Anemia/physiopathology , Coronary Vessels/physiology , Fetal Heart/physiology , Hyperemia/etiology , Microcirculation , Pregnancy Complications/physiopathology , Anemia/complications , Animals , Blood Pressure , Capillaries/physiology , Capillaries/physiopathology , Coronary Vessels/physiopathology , Female , Fetal Heart/physiopathology , Hyperemia/physiopathology , Pregnancy , Sheep
10.
FASEB J ; 29(10): 4346-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26139099

ABSTRACT

Studies in altricial rodents attribute dramatic changes in perinatal cardiomyocyte growth, maturation, and attrition to stimuli associated with birth. Our purpose was to determine whether birth is a critical trigger controlling perinatal cardiomyocyte growth, maturation and attrition in a precocial large mammal, sheep (Ovis aries). Hearts from 0-61 d postnatal lambs were dissected or enzymatically dissociated. Cardiomyocytes were measured by micromorphometry, cell cycle activity assessed by immunohistochemistry, and nuclear number counted after DNA staining. Integration of this new data with published fetal data from our laboratory demonstrate that a newly appreciated >30% decrease in myocyte number occurred in the last 10 d of gestation (P < 0.0005) concomitant with an increase in cleaved poly (ADP-ribose) polymerase 1 (P < 0.05), indicative of apoptosis. Bisegmental linear regressions show that most changes in myocyte growth kinetics occur before birth (median = 15.2 d; P < 0.05). Right ventricular but not left ventricular cell number increases in the neonate, by 68% between birth and 60 d postnatal (P = 0.028). We conclude that in sheep few developmental changes in cardiomyocytes result from birth, excepting the different postnatal degrees of free wall hypertrophy between the ventricles. Furthermore, myocyte number is reduced in both ventricles immediately before term, but proliferation increases myocyte number in the neonatal right ventricle.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Cell Proliferation , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Blotting, Western , Cell Count , Cell Size , Female , Fetus/cytology , Heart/growth & development , Linear Models , Male , Microscopy, Fluorescence , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Organ Size , Poly(ADP-ribose) Polymerases/metabolism , Pregnancy , Sheep , Time Factors
11.
Basic Clin Pharmacol Toxicol ; 117(4): 226-33, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25728792

ABSTRACT

Understanding theophylline pharmacokinetics (PK) in the foetus is essential to prevent in utero toxicity and optimize prophylactic therapies. Previous studies in pregnancy have been obfuscated by maternal dosing and inadequate sampling in the foetus; both render modelling of foetal PK difficult. Six ewes carrying singleton foetuses received theophylline (60 mg) into the foetal jugular vein. Blood samples were drawn from the foetus and ewe over 36 hr. Serum concentrations were measured. Maternal and foetal pharmacokinetic parameters were estimated. Foetal non-compartmental pharmacokinetic parameters were as follows: half-life 7.37 ± 1.22 hr; volume of distribution 44.62 ± 11.45 L; area under the curve 14.82 ± 2.71 hr/(µg/mL); and clearance 4.15 ± 0.70 L/hr. Rapid theophylline distribution across the placenta was observed. Maternal non-compartmental pharmacokinetic parameters were as follows: half-life 6.54 ± 2.44 hr; volume of distribution 32.48 ± 9.99 L; area under the curve 16.28 ± 4.53 hr/(µg/mL); and clearance 3.69 ± 1.47 L/hr. Foetal and ewe serum concentration-time profiles were fit together into a 3-compartment population pharmacokinetic model, and parameters were as follows: central volume 1.38 ± 0.11 L; 2nd peripheral compartment volume 3.11 ± 0.29 L; 3rd peripheral compartment volume 60.14 ± 6.02 L; elimination clearance 9.89 ± 0.90 L/hr; distribution clearance between central and 2nd compartment 30.87 ± 2.31 L/hr; and distribution clearance between 2nd and 3rd compartments 13.89 ± 1.11 L/hr. Cytochrome P4501A expression was robust in maternal liver; negligible activities were observed in placenta, foetal liver and foetal kidney. In vitro protein binding of theophylline was 30% lower in foetal serum compared to maternal serum (29.7 ± 4.4 versus 42.0 ± 3.6%-bound). Free concentrations were lower in the foetus than in the ewe, suggesting active transport across placenta. In summary, foetal clearance of theophylline is attributable to rapid distribution into the maternal circulation across the placenta followed by greater maternal protein binding and metabolic activity.


Subject(s)
Fetus/metabolism , Maternal-Fetal Exchange , Placental Circulation , Theophylline/pharmacokinetics , Animals , Area Under Curve , Female , Fetal Blood/metabolism , Half-Life , Injections, Intravenous , Metabolic Clearance Rate , Models, Biological , Pregnancy , Protein Binding , Risk Assessment , Sheep , Theophylline/administration & dosage , Theophylline/blood , Theophylline/toxicity
12.
Am J Physiol Regul Integr Comp Physiol ; 308(8): R680-9, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25652537

ABSTRACT

Hypoxia is a common component of many developmental insults and has been studied in early-stage chicken development. However, its impact on cardiac function and arterial-ventricular coupling in late-stage chickens is relatively unknown. To test the hypothesis that hypoxic incubation would reduce baseline cardiac function but protect the heart during acute hypoxia in late-stage chickens, white Leghorn eggs were incubated at 21% O2 or 15% O2. At 90% of incubation (19 days), hypoxic incubation caused growth restriction (-20%) and increased the LV-to-body ratio (+41%). Left ventricular (LV) pressure-volume loops were measured in anesthetized chickens in normoxia and acute hypoxia (10% O2). Hypoxic incubation lowered the maximal rate of pressure generation (ΔP/ΔtMax; -22%) and output (-57%), whereas increasing end-systolic elastance (ELV; +31%) and arterial elastance (EA; +122%) at similar heart rates to normoxic incubation. Both hypoxic incubation and acute hypoxia lengthened the half-time of relaxation (τ; +24%). Acute hypoxia reduced heart rate (-8%) and increased end-diastolic pressure (+35%). Hearts were collected for mRNA analysis. Hypoxic incubation was marked by decreased mRNA expression of sarco(endo)plasmic reticulum Ca(2+)-ATPase 2, Na(+)/Ca(2+) exchanger 1, phospholamban, and ryanodine receptor. In summary, hypoxic incubation reduces LV function in the late-stage chicken by slowing pressure generation and relaxation, which may be driven by altered intracellular excitation-contraction coupling. Cardiac efficiency is greatly reduced after hypoxic incubation. In both incubation groups acute hypoxia reduced diastolic function.


Subject(s)
Cardiac Catheterization , Heart/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Hypoxia/physiopathology , Stroke Volume , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Pressure , Animals , Chick Embryo , Chronic Disease , Disease Models, Animal , Excitation Contraction Coupling , Gene Expression Regulation, Developmental , Heart/embryology , Hypertrophy, Left Ventricular/embryology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypoxia/embryology , Hypoxia/genetics , Hypoxia/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Myocardial Contraction , RNA, Messenger/metabolism , Stroke Volume/genetics , Time Factors , Ventricular Dysfunction, Left/embryology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left/genetics , Ventricular Pressure/genetics
13.
Am J Physiol Heart Circ Physiol ; 305(10): H1530-7, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24014675

ABSTRACT

Obesity increases the risk of arrhythmias and sudden cardiac death, but the mechanisms are unknown. This study tested the hypothesis that obesity-induced cardiac sympathetic outgrowth and hyperinnervation promotes the development of arrhythmic events. Male Sprague-Dawley rats (250-275 g), fed a high-fat diet (33% kcal/fat), diverged into obesity-resistant (OR) and obesity-prone (OP) groups and were compared with rats fed normal chow (13% kcal/fat; CON). In vitro experiments showed that both OR and OP rats exhibited hyperinnervation of the heart and high sympathetic outgrowth compared with CON rats, even though OR rats are not obese. Despite the hyperinnervation and outgrowth, we showed that, in vivo, OR rats were less susceptible to arrhythmic events after an intravenous epinephrine challenge compared with OP rats. On examining total and stimulus-evoked neurotransmitter levels in an ex vivo system, we demonstrate that atrial acetylcholine content and release were attenuated in OP compared with OR and CON groups. OP rats also expressed elevated atrial norepinephrine content, while norepinephrine release was suppressed. These findings suggest that the consumption of a high-fat diet, even in the absence of overt obesity, stimulates sympathetic outgrowth and hyperinnervation of the heart. However, normalized cardiac parasympathetic nervous system control may protect the heart from arrhythmic events.


Subject(s)
Arrhythmias, Cardiac/etiology , Diet, High-Fat , Heart/innervation , Obesity/etiology , Sympathetic Nervous System/physiopathology , Acetylcholine/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Disease Models, Animal , Epinephrine , Heart Atria/innervation , Male , Norepinephrine/metabolism , Obesity/complications , Obesity/metabolism , Obesity/physiopathology , Parasympathetic Nervous System/metabolism , Parasympathetic Nervous System/physiopathology , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/metabolism
15.
Reprod Sci ; 19(6): 642-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22421446

ABSTRACT

Circulating fetal 3,3',5-tri-iodo-l-thyronine (T(3) ) is maintained at very low levels until a dramatic prepartum surge. 3,3',5-Tri-iodo-l-thyronine inhibits serum-stimulated proliferation in near-term ovine cardiomyocytes, but it is not known whether midgestation myocytes are also inhibited. Because early cessation of cardiomyocyte mitosis would result in an underendowed heart, we hypothesized that 0.67 gestation (100 of 145 days gestation) ovine cardiomyocytes would be insensitive to suppressive growth effects of T(3) . These younger cardiomyocytes were grown with T(3) in 10% serum-enriched media for 24 hours. Physiological (0.37, 0.75, and 1.5 nmol/L) concentrations of T(3) dramatically suppressed mitotic activity in cardiomyocytes (P < .001). 3,3',5-Tri-iodo-l-thyronine stimulated phosphorylation of extracellular signal-regulated kinase and AKT (also known as Protein Kinase B [PKB]) signaling pathways. Nevertheless, the protein content of the cell cycle suppressor, p21, increased 2-fold (P < .05), and promoter, cyclin D1, decreased by 50%. Contrary to our hypothesis, elevated levels of T(3) powerfully inhibit proliferation of midgestation fetal cardiomyocytes. Thus, midgestation maternal hyperthyroidism might lead to an underendowed fetal myocardium.


Subject(s)
Fetal Heart/cytology , Mitosis/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Sheep/embryology , Thyroid Hormones/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/analysis , Cyclin-Dependent Kinase Inhibitor p21/analysis , Female , Gestational Age , Myocytes, Cardiac/chemistry , Pregnancy , Signal Transduction/drug effects , Triiodothyronine/pharmacology
16.
FASEB J ; 26(1): 397-408, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21974928

ABSTRACT

Tri-iodo-l-thyronine (T(3)) suppresses the proliferation of near-term serum-stimulated fetal ovine cardiomyocytes in vitro. Thus, we hypothesized that T(3) is a major stimulant of cardiomyocyte maturation in vivo. We studied 3 groups of sheep fetuses on gestational days 125-130 (term ∼145 d): a T(3)-infusion group, to mimic fetal term levels (plasma T(3) levels increased from ∼0.1 to ∼1.0 ng/ml; t(1/2)∼24 h); a thyroidectomized group, to produce low thyroid hormone levels; and a vehicle-infusion group, to serve as intact controls. At 130 d of gestation, sections of left ventricular freewall were harvested, and the remaining myocardium was enzymatically dissociated. Proteins involved in cell cycle regulation (p21, cyclin D1), proliferation (ERK), and hypertrophy (mTOR) were measured in left ventricular tissue. Evidence that elevated T(3) augmented the maturation rate of cardiomyocytes included 14% increased width, 31% increase in binucleation, 39% reduction in proliferation, 150% reduction in cyclin D1 protein, and 500% increase in p21 protein. Increased expression of phospho-mTOR, ANP, and SERCA2a also suggests that T(3) promotes maturation and hypertrophy of fetal cardiomyocytes. Thyroidectomized fetuses had reduced cell cycle activity and binucleation. These findings support the hypothesis that T(3) is a prime driver of prenatal cardiomyocyte maturation.


Subject(s)
Heart/embryology , Heart/physiology , Myocytes, Cardiac/physiology , Triiodothyronine/physiology , Animals , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cyclin D1/physiology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , Gestational Age , Hemodynamics/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Organ Size , Pregnancy , Sheep , Thyroidectomy , Triiodothyronine/deficiency , Triiodothyronine/pharmacology
17.
Am J Physiol Heart Circ Physiol ; 298(6): H1652-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20190098

ABSTRACT

Sympathetic nerves stimulate cardiac function through the release of norepinephrine and the activation of cardiac beta(1)-adrenergic receptors. The sympathetic innervation of the heart is sculpted during development by chemoattractive factors including nerve growth factor (NGF) and the chemorepulsive factor semaphorin 3a. NGF acts through the TrkA receptor and the p75 neurotrophin receptor (p75(NTR)) in sympathetic neurons. NGF stimulates sympathetic axon extension into the heart through TrkA, but p75(NTR) modulates multiple coreceptors that can either stimulate or inhibit axon outgrowth. In mice lacking p75(NTR), the sympathetic innervation density in target tissues ranges from denervation to hyperinnervation. Recent studies have revealed significant changes in the sympathetic innervation density of p75NTR-deficient (p75(NTR-/-)) atria between early postnatal development and adulthood. We examined the innervation of adult p75(NTR-/-) ventricles and discovered that the subendocardium of the p75(NTR-/-) left ventricle was essentially devoid of sympathetic nerve fibers, whereas the innervation density of the subepicardium was normal. This phenotype is similar to that seen in mice overexpressing semaphorin 3a, and we found that sympathetic axons lacking p75(NTR) are more sensitive to semaphorin 3a in vitro than control neurons. The lack of subendocardial innervation was associated with decreased dP/dt, altered cardiac beta(1)-adrenergic receptor expression and sensitivity, and a significant increase in spontaneous ventricular arrhythmias. The lack of p75(NTR) also resulted in increased tyrosine hydroxylase content in cardiac sympathetic neurons and elevated norepinephrine in the right ventricle, where innervation density was normal.


Subject(s)
Arrhythmias, Cardiac/metabolism , Heart Ventricles/innervation , Receptors, Adrenergic, beta-1/metabolism , Receptors, Nerve Growth Factor/metabolism , Sympathetic Nervous System/growth & development , Ventricular Dysfunction/metabolism , Animals , Arrhythmias, Cardiac/physiopathology , Disease Models, Animal , Female , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Norepinephrine/metabolism , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/genetics , Semaphorin-3A/metabolism , Signal Transduction/physiology , Ventricular Dysfunction/physiopathology
18.
Exp Physiol ; 95(1): 131-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19700519

ABSTRACT

Chronic anaemia increases the workload of the growing fetal heart, leading to cardiac enlargement. To determine which cellular process increases cardiac mass, we measured cardiomyocyte sizes, binucleation as an index of terminal differentiation, and tissue volume fractions in hearts from control and anaemic fetal sheep. Fourteen chronically catheterized fetal sheep at 129 days gestation had blood withdrawn for 9 days to cause severe anaemia; 14 control fetuses were of similar age. At postmortem examination, hearts were either enzymatically dissociated or fixed for morphometric analysis. Daily isovolumetric haemorrhage reduced fetal haematocrit from a baseline value of 35% to 15% on the final day (P < 0.001). At the study conclusion, anaemic fetuses had lower arterial pressures than control fetuses (P < 0.05). Heart weights were increased by 39% in anaemic fetuses compared with control hearts (P < 0.0001), although the groups had similar body weights; the heart weight difference was not due to increased ventricular wall water content or disproportionate non-myocyte tissue expansion. Cardiomyocytes from anaemic fetuses tended to be larger than those of control fetuses. There were no statistically significant differences between groups in the cardiomyocyte cell cycle activity. The degree of terminal differentiation was greater in the right ventricle of anaemic compared with control fetuses by 8% (P < 0.05). Anaemia substantially increased heart weight in fetal sheep. The volume proportions of connective and vascular tissue were unchanged. Cardiomyocyte mass expanded by a balanced combination of cellular enlargement, increased terminal differentiation and accelerated proliferation.


Subject(s)
Anemia/pathology , Cell Enlargement , Cell Proliferation , Disease Models, Animal , Fetal Diseases/pathology , Myocytes, Cardiac/pathology , Anemia/blood , Animals , Chronic Disease , Female , Fetal Diseases/blood , Myocytes, Cardiac/metabolism , Pregnancy , Sheep
19.
Respir Physiol Neurobiol ; 165(1): 22-7, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18952009

ABSTRACT

The nose conditions the temperature and humidity of nasal air, and the nasal mucosal vasculature supplies heat and water for these processes. We hypothesize that nitric oxide (NO) modulates these processes through vasoactive effects on nasal mucosal vasculature. We measured the temperature, humidity and NO concentrations of nasal air during inhalation and exhalation across the nose and calculated net heat, water and NO output before (controls, n=7) and after inhibition of NO synthase by topical l-NAME (N=5) in healthy humans. We found that calculated NO output across the nasal passages is approximately three-fold greater during inhalation (503+/-105 nL/min) compared with exhalation (162+/-56 nL/min). Moreover, topical administration of l-NAME decreased nasal air temperature and humidity conditioning and NO output, but these effects were limited to inhalation. We conclude that nasal NO output is greater during inhalation than exhalation in humans. Our findings also support a role of nasal NO in temperature and humidity conditioning of nasal air.


Subject(s)
Air , Nitric Oxide/metabolism , Nose/physiology , Respiratory Mechanics/physiology , Temperature , Water , Adult , Body Temperature/drug effects , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Middle Aged , NG-Nitroarginine Methyl Ester/pharmacology , Nose/drug effects , Plethysmography/methods , Respiratory Mechanics/drug effects , Young Adult
20.
Article in English | MEDLINE | ID: mdl-18196943

ABSTRACT

While it is established that the quality of the perinatal environment is critical in sculpting the developing individual, the mechanisms by which this occurs remain poorly defined. The growing fetus is dependent on the nutrients (including oxygen) it receives from the mother via the placenta. When this supply line is compromised, heart growth patterns are altered. In addition, hormones, other circulating factors, and the hemodynamic environment in which the fetus develops are important in determining outcomes for organ structure and function. Numerous studies in sheep have demonstrated that heart development can be modified in a number of ways, and the nature of the change differs between types and gestational timings of insults. Embolization of the placenta leads to the cessation of proliferation and maturation of cardiomyocytes; this may be due in part to changes in circulating insulin-like growth factor-1 levels. Such insults may be the underlying cause of cardiovascular disease in adults. Insults that modify the maturational timeline, final myocyte number, vascularity and endothelial responsiveness in the heart can have effects that persist long after the insult has been ameliorated.


Subject(s)
Heart , Maternal Nutritional Physiological Phenomena/physiology , Placenta/physiology , Pregnancy/physiology , Prenatal Nutritional Physiological Phenomena/physiology , Female , Gestational Age , Heart/embryology , Heart/growth & development , Heart/physiology , Humans , Insulin-Like Growth Factor I/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Nutrition Disorders/physiopathology , Placenta/blood supply
SELECTION OF CITATIONS
SEARCH DETAIL
...