Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Hypertens Res ; 47(7): 1897-1907, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38664509

ABSTRACT

Artificial light at night (ALAN) disrupts 24-h variability of blood pressure, but the molecular mechanisms underlying these effects are unknown. Therefore, we analysed the daily variability of pulse pressure, the maximum value of acceleration rate of aortic pressure (dP/dt(max)) measured by telemetry and protein expression in the thoracic aorta of normotensive male rats exposed to ALAN (1-2 lx) for 3 weeks. Daily, 24-h variability of pulse pressure and dP/dt(max) was observed during a regular light/dark regimen with higher values during the dark compared to the light phase of the day. ALAN suppressed 24-h variability and enhanced ultradian (<12-h) periods of pulse pressure and dP/dt(max) in duration-dependent manners. From beat-to-beat blood pressure variability, ALAN decreased low-frequency bands (a sympathetic marker) and had minimal effects on high-frequency bands. At the molecular level, ALAN decreased angiotensin II receptor type 1 expression and reduced 24-h variability. ALAN caused the appearance of 12-h oscillations in transforming growth factor ß1 and fibulin 4. Expression of sarco/endoplasmic reticulum Ca2+-ATPase type 2 was increased in the middle of the light and dark phase of the day, and ALAN did not affect its daily and 12-h variability. In conclusion, ALAN suppressed 24-h variability of pulse pressure and dP/dt(max), decreased the power of low-frequency bands and differentially affected the expression of specific proteins in the rat thoracic aorta. Suppressed 24-h oscillations by ALAN underline the pulsatility of individual endocrine axes with different periods, disrupting the cardiovascular control of central blood pressure.


Subject(s)
Aorta, Thoracic , Blood Pressure , Circadian Rhythm , Animals , Male , Aorta, Thoracic/metabolism , Blood Pressure/physiology , Rats , Circadian Rhythm/physiology , Transforming Growth Factor beta1/metabolism , Receptor, Angiotensin, Type 1/metabolism , Light , Extracellular Matrix Proteins/metabolism , Rats, Sprague-Dawley , Calcium-Binding Proteins/metabolism
3.
Pflugers Arch ; 476(3): 295-306, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38177874

ABSTRACT

Artificial light at night (ALAN) affects most of the population. Through the retinohypothalamic tract, ALAN modulates the activity of the central circadian oscillator and, consequently, various physiological systems, including the cardiovascular one. We summarised the current knowledge about the effects of ALAN on the cardiovascular system in diurnal and nocturnal animals. Based on published data, ALAN reduces the day-night variability of the blood pressure and heart rate in diurnal and nocturnal animals by increasing the nocturnal values of cardiovascular variables in diurnal animals and decreasing them in nocturnal animals. The effects of ALAN on the cardiovascular system are mainly transmitted through the autonomic nervous system. ALAN is also considered a stress-inducing factor, as glucocorticoid and glucose level changes indicate. Moreover, in nocturnal rats, ALAN increases the pressure response to load. In addition, ALAN induces molecular changes in the heart and blood vessels. Changes in the cardiovascular system significantly depend on the duration of ALAN exposure. To some extent, alterations in physical activity can explain the changes observed in the cardiovascular system after ALAN exposure. Although ALAN acts differently on nocturnal and diurnal animals, we can conclude that both exhibit a weakened circadian coordination among physiological systems, which increases the risk of future cardiovascular complications and reduces the ability to anticipate stress.


Subject(s)
Cardiovascular System , Light , Humans , Rats , Animals , Light Pollution , Blood Pressure , Heart Rate
4.
Life Sci ; 333: 122179, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37852575

ABSTRACT

AIMS: Early postnatal development can be significantly compromised by changes in factors provided by the mother, leading to increased vulnerability to hypertension in her offspring. TGR(mRen-2)27 (TGR) mothers, characterised by an overactivated renin-angiotensin system, exhibit altered ion composition in their breast milk. Therefore, we aimed to analyse the impact of cross-fostering on cardiovascular parameters in hypertensive TGR and normotensive Hannover Sprague-Dawley (HanSD) offspring. MATERIALS AND METHODS: We measured cardiovascular parameters in 5- to 10-week-old male offspring by telemetry. The expression of proteins related to vascular function was assessed by western blotting in the aortic samples obtained from 6- to 12-week-old male offspring. Plasma renin activity and plasma angiotensin II (Ang II) levels were evaluated by radioimmunoassay (RIA). KEY FINDINGS: The development of hypertension was in TGR accompanied by increased low-to-high frequency ratio (LF/HF; a marker of sympathovagal balance; 0.51 ± 0.16 in week 10). Furthermore, TGR exhibited increased aortic expression of mineralocorticoid receptor (MR; p < 0.05) and transforming growth factor beta type 1 (TGF-ß1; p = 0.002) compared to HanSD offspring. Fostering significantly decreased sympathovagal balance (0.23 ± 0.10 in week 10) and, transiently, plasma Ang II levels and MR expression in TGR offspring reared by HanSD mothers. SIGNIFICANCE: These findings highlight the importance of understanding the complex interplay between early life experiences, maternal factors, and later cardiovascular function. Understanding the mechanisms behind the observed effects may help to identify potential interventions to prevent the development of hypertension later in life.


Subject(s)
Hypertension , Kidney , Humans , Female , Animals , Rats , Male , Animals, Genetically Modified , Kidney/metabolism , Mothers , Renin , Rats, Sprague-Dawley , Blood Pressure/physiology , Angiotensin II/metabolism
5.
Mol Cell Endocrinol ; 572: 111967, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37210027

ABSTRACT

Aldosterone regulates blood pressure (BP) through water and sodium balance. In our study, we studied if continuous treatment with a mineralocorticoid receptor antagonist, spironolactone (30 mg/kg/day) for 20 days can: 1) attenuate hypertension development and restore inverted 24-h BP rhythm in hypertensive transgenic (mRen-2)27 rats (TGR) measured by telemetry; 2) improve function of the kidneys and heart; 3) be protective against high salt load (1% in water) by mitigating oxidative injury and improving kidney function. Spironolactone decreased albuminuria and 8-isoprostane in normal and salt load conditions in BP-independent effects. Salt load increased BP, impaired autonomic balance, suppressed plasma aldosterone level and increased natriuresis, albuminuria and oxidative injury in TGR. Spironolactone did not restore the inverted 24-h rhythm of BP in TGR, therefore, mineralocorticoids are not crucial in regulation of BP daily profile. Spironolactone improved kidney function, decreased oxidative stress and was protective against high salt load in the BP-independent manner.


Subject(s)
Aldosterone , Hypertension , Rats , Animals , Blood Pressure , Aldosterone/pharmacology , Receptors, Mineralocorticoid/genetics , Spironolactone/pharmacology , Albuminuria , Kidney , Animals, Genetically Modified , Water/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology
6.
Hypertens Res ; 45(12): 1929-1944, 2022 12.
Article in English | MEDLINE | ID: mdl-36123396

ABSTRACT

The hypotensive effects of melatonin are based on a negative correlation between melatonin levels and blood pressure in humans. However, there is a positive correlation in nocturnal animals that are often used as experimental models in cardiovascular research, and the hypotensive effects and mechanism of melatonin action are often investigated in rats and mice. In rats, the hypotensive effects of melatonin have been studied in normotensive and spontaneously or experimentally induced hypertensive strains. In experimental animals, blood pressure is often measured indirectly during the light (passive) phase of the day by tail-cuff plethysmography, which has limitations regarding data quality and animal well-being compared to telemetry. Melatonin is administered to rats in drinking water, subcutaneously, intraperitoneally, or microinjected into specific brain areas at different times. Experimental data show that the hypotensive effects of melatonin depend on the experimental animal model, blood pressure measurement technique, and the route, time and duration of melatonin administration. The hypotensive effects of melatonin may be mediated through specific membrane G-coupled receptors located in the heart and arteries. Due to melatonin's lipophilic nature, its potential hypotensive effects can interfere with various regulatory mechanisms, such as nitric oxide and reactive oxygen species production and activation of the autonomic nervous and circadian systems. Based on the research conducted on rats, the cardiovascular effects of melatonin are modulatory, delayed, and indirect.


Subject(s)
Cardiovascular System , Hypertension , Hypotension , Melatonin , Humans , Rats , Animals , Mice , Melatonin/pharmacology , Melatonin/therapeutic use , Melatonin/physiology , Blood Pressure , Hypertension/drug therapy
7.
Int J Mol Sci ; 23(5)2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35270026

ABSTRACT

Prenatal hypoxia during the prenatal period can interfere with the developmental trajectory and lead to developing hypertension in adulthood. Prenatal hypoxia is often associated with intrauterine growth restriction that interferes with metabolism and can lead to multilevel changes. Therefore, we analysed the effects of prenatal hypoxia predominantly not associated with intrauterine growth restriction using publications up to September 2021. We focused on: (1) The response of cardiovascular regulatory mechanisms, such as the chemoreflex, adenosine, nitric oxide, and angiotensin II on prenatal hypoxia. (2) The role of the placenta in causing and attenuating the effects of hypoxia. (3) Environmental conditions and the mother's health contribution to the development of prenatal hypoxia. (4) The sex-dependent effects of prenatal hypoxia on cardiovascular regulatory mechanisms and the connection between hypoxia-inducible factors and circadian variability. We identified that the possible relationship between the effects of prenatal hypoxia on the cardiovascular regulatory mechanism may vary depending on circadian variability and phase of the days. In summary, even short-term prenatal hypoxia significantly affects cardiovascular regulatory mechanisms and programs hypertension in adulthood, while prenatal programming effects are not only dependent on the critical period, and sensitivity can change within circadian oscillations.


Subject(s)
Cardiovascular System , Hypertension , Prenatal Exposure Delayed Effects , Adult , Female , Fetal Growth Retardation , Humans , Hypoxia/complications , Pregnancy
8.
Physiol Behav ; 242: 113620, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34637804

ABSTRACT

Cardiovascular (CV) health is often expressed by changes in heart rate and blood pressure, the physiological record of which may be affected by moving, anaesthesia, handling, time of day and many other factors in rodents. Telemetry measurement minimises these modulations and enables more accurate physiological recording of heart rate and blood pressure than non-invasive methods. Measurement of arterial blood pressure by telemetry requires implanting a catheter tip into the artery. Telemetry enables us to sample physiological parameters with a high frequency continuously for several months. By measuring the pressure in the artery using telemetry, we can visualize pressure changes over a heart cycle as the pressure wave. From the pressure wave, we can subtract systolic, diastolic, mean and pulse pressure. From the beat-to-beat interval (pressure wave) and the RR' interval (electrocardiogram), we can derive the heart rate. From beat-to-beat variability, we can evaluate the autonomic nervous system's activity and spontaneous baroreflex sensitivity and their impact on CV activity. On a long-term scale, circadian variability of CV parameters is evident. Circadian variability is the result of the circadian system's activity, which synchronises and organises many activities in the body, such as autonomic and reflex modulation of the CV system and its response to load over the day. In the presented review, we aimed to discuss telemetry devices, their types, implantation, set-up, limitations, short-term and long-term variability of heart rate and blood pressure in CV research. Data collection by telemetry should be, despite some limitations, standard in modern experimental CV research.


Subject(s)
Baroreflex , Telemetry , Blood Pressure , Data Collection , Heart Rate
9.
Exp Physiol ; 106(8): 1762-1771, 2021 08.
Article in English | MEDLINE | ID: mdl-34089548

ABSTRACT

NEW FINDINGS: What is the central question of this study? Artificial light at night decreases blood pressure and heart rate in rats. Are these changes in heart rate accompanied by changes in protein expression in the heart's left ventricle? What is the main finding and its importance? Five weeks of artificial light at night affected protein expression in the heart's left ventricle in normotensive and hypertensive rats. Artificial light at night decreased expression of the sarco/endoplasmic reticulum Ca2+ -ATPase, angiotensin II receptor type 1 and endothelin-1. ABSTRACT: Artificial light at night (ALAN) affects the circadian rhythm of the heart rate in normotensive Wistar rats (WT) and spontaneously hypertensive rats (SHR) through the autonomic nervous system, which regulates the heart's activity through calcium handling, an important regulator in heart contractility. We analysed the expression of the sarco/endoplasmic reticulum Ca2+ -ATPase (SERCA2) and other selected regulatory proteins involved in the regulation of heart contractility, angiotensin II receptor type 1 (AT1 R), endothelin-1 (ET-1) and tyrosine hydroxylase (TH), in the left ventricle of the heart in WT and SHR after 2 and 5 weeks of ALAN with intensity 1-2 lx. Expression of SERCA2 was decreased in WT (control: 0.53 ± 0.07; ALAN: 0.46 ± 0.10) and SHR (control: 0.72 ± 0.18; ALAN: 0.56 ± 0.21) after 5 weeks of ALAN (P = 0.067). Expression of AT1 R was significantly decreased in WT (control: 0.51 ± 0.27; ALAN: 0.34 ± 0.20) and SHR (control: 0.38 ± 0.07; ALAN: 0.23 ± 0.09) after 2 weeks of ALAN (P = 0.028) and in SHR after 5 weeks of ALAN. Expression of ET-1 was decreased in WT (control: 0.51 ± 0.27; ALAN: 0.28 ± 0.12) and SHR (control: 0.54 ± 0.10; ALAN: 0.35 ± 0.23) after 5 weeks of ALAN (P = 0.015). ALAN did not affect the expression of TH in WT or SHR. In conclusion, ALAN suppressed the expression of SERCA2, AT1 R and ET-1, which are important for the regulation of heart contractility, in a strain-dependent pattern in both WT and SHR.


Subject(s)
Heart Ventricles , Hypertension , Animals , Blood Pressure , Endoplasmic Reticulum/metabolism , Light Pollution , Rats , Rats, Wistar
10.
J Dev Orig Health Dis ; 12(4): 587-594, 2021 08.
Article in English | MEDLINE | ID: mdl-33109302

ABSTRACT

Prenatal hypoxia (PH) has negative consequences on the cardiovascular system in adulthood and can affect the responses to additional insults later in life. We explored the effects of PH imposed during embryonic day 20 (10.5% O2 for 12 h) on circadian rhythms of systolic blood pressure (BP) and heart rate (HR) in mature male rat offspring measured by telemetry. We evaluated: (1) stability of BP and HR changes after PH; (2) circadian variability of BP and HR after 2 and 5 weeks of exposure to artificial light at night (ALAN; 1-2 lx); and (3) response of BP and HR to norepinephrine. PH increased BP in the dark (134 ± 2 mmHg vs. control 127 ± 2 mmHg; p = 0.05) and marginally in the light (125 ± 1 mmHg vs. control 120 ± 2 mmHg) phase of the day but not HR. The effect of PH was highly repeatable between 21- and 27-week-old PH male offspring. Two weeks of ALAN decreased the circadian variability of HR (p < 0.05) and BP more in control than PH rats. After 5 weeks of ALAN, the circadian variability of HR and BP were damped compared to LD and did not differ between control and PH rats (p < 0.05). Responses of BP and HR to norepinephrine did not differ between control and PH rats. Hypoxia at the end of the embryonic period increases BP and affects the functioning of the cardiovascular system in mature male offspring. ALAN in adulthood decreased the circadian variability of cardiovascular parameters, more in control than PH rats.


Subject(s)
Blood Pressure , Circadian Rhythm , Heart Rate , Hypoxia/physiopathology , Prenatal Exposure Delayed Effects , Animals , Female , Light , Male , Pregnancy , Rats, Wistar
11.
Cell Mol Neurobiol ; 41(7): 1589-1598, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32734322

ABSTRACT

Melatonin is released by the pineal gland and can modulate cardiovascular system function via the G protein-coupled melatonin receptors MT1 and MT2. Most vessels are surrounded by perivascular adipose tissue (PVAT), which affects their contractility. The aim of our study was to evaluate mRNA and protein expression of MT1 and MT2 in the mesenteric artery (MA) and associated PVAT of male rats by RT-PCR and Western blot. Receptor localization was further studied by immunofluorescence microscopy. Effects of melatonin on neurogenic contractions were explored in isolated superior MA ex vivo by measurement of isometric contractile tension. MT1, but not MT2, was present in MA, and MT1 was localized mainly in vascular smooth muscle. Moreover, we proved the presence of MT1, but not MT2 receptors, in MA-associated PVAT. In isolated superior MA with intact PVAT, neuro-adrenergic contractile responses were significantly smaller when compared to arteries with removed PVAT. Pre-treatment with melatonin of PVAT-stripped arterial rings enhanced neurogenic contractions, while the potentiating effect of melatonin was not detected in preparations with preserved PVAT. We hypothesize that melatonin can stimulate the release of PVAT-derived relaxing factor(s) via MT1, which can override the direct pro-contractile effect of melatonin on vascular smooth muscle. Our results suggest that melatonin is involved in the control of vascular tone in a complex way, which is vessel specific and can reflect a sum of action on different layers of the vessel wall and surrounding PVAT.


Subject(s)
Melatonin/pharmacology , Mesenteric Arteries/metabolism , Receptors, Melatonin/drug effects , Receptors, Melatonin/metabolism , Adipose Tissue/drug effects , Adipose Tissue/physiology , Animals , Melatonin/metabolism , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Rats, Wistar
12.
Life Sci ; 231: 116568, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31202842

ABSTRACT

AIMS: Cardiovascular parameters exhibit significant 24-h variability, which is coordinated by the suprachiasmatic nucleus (SCN), and light/dark cycles control SCN activity. We aimed to study the effects of light at night (ALAN; 1-2 lx) on cardiovascular system control in normotensive rats. MAIN METHODS: Heart rate (HR) and blood pressure (BP) were measured by telemetry during five weeks of ALAN exposure. From beat-to-beat telemetry data, we evaluated spontaneous baroreflex sensitivity (sBRS). After 2 (A2) and 5 (A5) weeks of ALAN, plasma melatonin concentrations and the response of BP and HR to norepinephrine administration were measured. The expression of endothelial nitric oxide synthase (eNOS) and endothelin-1 was determined in the aorta. Spontaneous exploratory behaviour was evaluated in an open-field test. KEY FINDINGS: ALAN significantly suppressed the 24-h variability in the HR, BP, and sBRS after A2, although the parameters were partially restored after A5. The daily variability in the BP response to norepinephrine was reduced after A2 and restored after A5. ALAN increased the BP response to norepinephrine compared to the control after A5. Increased eNOS expression was found in arteries after A2 but not A5. Endothelin-1 expression was not affected by ALAN. Plasma melatonin levels were suppressed after A2 and A5. Spontaneous exploratory behaviour was reduced. SIGNIFICANCE: ALAN decreased plasma melatonin and the 24-h variability in the haemodynamic parameters and increased the BP response to norepinephrine. A low intensity ALAN can suppress circadian control of the cardiovascular system with negative consequences on the anticipation of a load.


Subject(s)
Circadian Rhythm/physiology , Lighting/methods , Melatonin/metabolism , Animals , Baroreflex , Blood Pressure , Cardiovascular System/metabolism , Heart Rate , Hemodynamics , Light , Male , Norepinephrine/metabolism , Rats , Rats, Wistar , Suprachiasmatic Nucleus/metabolism
13.
Can J Physiol Pharmacol ; 97(9): 863-871, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31251886

ABSTRACT

Circadian rhythms are an inherent property of physiological processes and can be disturbed by irregular environmental cycles, including artificial light at night (ALAN). Circadian disruption may contribute to many pathologies, such as hypertension, obesity, and type 2 diabetes, but the underlying mechanisms are not understood. Our study investigated the consequences of ALAN on cardiovascular and metabolic parameters in spontaneously hypertensive rats, which represent an animal model of essential hypertension and insulin resistance. Adult males were exposed to a 12 h light - 12 h dark cycle and the ALAN group experienced dim light at night (1-2 lx), either for 2 or 5 weeks. Rats on ALAN showed a loss of light-dark variability for systolic blood pressure, but not for heart rate. Moreover, a gradual increase of systolic blood pressure was recorded over 5 weeks of ALAN. Exposure to ALAN increased plasma insulin and hepatic triglyceride levels. An increased expression of metabolic transcription factors, Pparα and Pparγ, in the epididymal fat and a decreased expression of Glut4 in the heart was found in the ALAN group. Our results demonstrate that low-intensity ALAN can disturb blood pressure control and augment insulin resistance in spontaneously hypertensive rats, and may represent a serious risk factor for cardiometabolic diseases.


Subject(s)
Blood Pressure/radiation effects , Circadian Rhythm/physiology , Circadian Rhythm/radiation effects , Heart Rate/radiation effects , Light/adverse effects , Animals , Blood Pressure/physiology , Dose-Response Relationship, Radiation , Heart Rate/physiology , Insulin/blood , Insulin Resistance/physiology , Insulin Resistance/radiation effects , Leptin/blood , Male , Rats , Rats, Inbred SHR
14.
Gen Physiol Biophys ; 38(3): 265-270, 2019 May.
Article in English | MEDLINE | ID: mdl-31184313

ABSTRACT

This study investigated the effect of lisinopril (angiotensin-converting enzyme inhibitor) on potential behavioural alterations in spontaneously hypertensive rats (SHR). Three groups of 15-17-week-old rats were investigated for 2 weeks: Wistar control group, SHR group and SHR+lisinopril group. Systolic blood pressure (SBP) was normal in Wistar rats, SHR expressed hypertension and lisinopril normalized the SBP. We observed increased time spent in and increased frequency of entries to the central area of the open field in SHR, while lisinopril induced a trend to reduce the time spent in the central area of the open field and reduced the frequency of entries there. There was a positive correlation between SBP and reduced anxiety-like behaviour in normotensive rats; no correlations in the SHR or SHR+lisinopril groups were observed. We conclude that lisinopril normalized the increase in SBP and partly reversed the alterations of anxiety-like behaviour in SHR.


Subject(s)
Antihypertensive Agents/pharmacology , Anxiety/drug therapy , Behavior, Animal/drug effects , Hypertension/drug therapy , Hypertension/psychology , Lisinopril/pharmacology , Animals , Anxiety/prevention & control , Blood Pressure , Hypertension/physiopathology , Rats , Rats, Inbred SHR , Rats, Wistar
15.
Nurs Crit Care ; 24(5): 320-325, 2019 09.
Article in English | MEDLINE | ID: mdl-31087602

ABSTRACT

BACKGROUND: Intensive care unit (ICU) patients experience two affronts to normal 24-h rhythms: largely internal events such as medication and external factors such as light, noise and nursing interventions. AIMS AND OBJECTIVES: We investigated the impact of light variance within an ICU on 24-h rhythmicity of three key physiological parameters: heart rate (HR), mean arterial blood pressure (MAP) and body temperature (BT) in this patient population. DESIGN: Patients were assigned to beds either in the 'light' or 'dark' side within a single ICU. An actigraph continuously recorded light intensity for a 24-72-h period. METHODS: Measurements of HR, MAP and BT were recorded every 30 min. RESULTS: HR, MAP and BT did not follow 24-h rhythmicity in all patients. Higher light exposure in the Light Side of the ICU (122·3 versus 50·6 lx) was related to higher HR (89·4 versus 79·8 bpm), which may translate to clinically relevant outcomes in a larger sample. Duration of stay, the one clinical outcome measured in this study, showed no significant variation between the groups (p = 0·147). CONCLUSIONS: ICU patients are exposed to varying light intensities depending on bed positioning relative to natural sunlight, affecting the 24-h rhythm of HR. Larger, well-controlled studies also investigating the effect of relevant light intensity are indicated. RELEVANCE TO CLINICAL PRACTICE: Light is a variable that can be manipulated in the constrained environment of an ICU, thus offering an avenue for relatively unobtrusive interventions.


Subject(s)
Arterial Pressure/physiology , Body Temperature/physiology , Circadian Rhythm/physiology , Critical Care Nursing/methods , Heart Rate/physiology , Light , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged
16.
Auton Neurosci ; 206: 51-59, 2017 09.
Article in English | MEDLINE | ID: mdl-28765031

ABSTRACT

Physiological variables such as heart rate (HR) and blood pressure (BP) exhibit long-term circadian rhythms, which can be disturbed by shift work. On the other hand, short-term oscillations in HR and BP have a high prognostic value. Therefore, we aimed to determine if the short-term variability, complexity and entropy of HR and BP would be affected by a regular light/dark (LD) cycle and phase delay shifts of the LD cycle, leading to chronodisruption. Telemetry-monitored rats were exposed first to the regular LD cycle and then to shifts in LD for 8weeks. On the basis of long-term HR and BP recording and evaluation, we found circadian rhythms in HR and BP variability, complexity and entropy under regular LD cycles. Short-term exposure to shifts disturbed circadian rhythms of HR and BP variability, complexity and entropy, indicating chronodisruption. The power of circadian rhythms was suppressed after 8weeks of phase delay shifts. Long-term exposure to shifts increased variability (p=0.007), complexity (p<0.001) and dark-time entropy (p=0.006) of HR but not BP. This is the first study demonstrating long-term recording and estimation of HR and BP variability, complexity and entropy in conscious rats exposed to irregular lighting conditions. After long-term phase delay shifts, short-term variability of HR was less predictable than in controls. This study suggests that changes in short-term HR and BP oscillations induced by long-term shift work can negatively affect cardiovascular health.


Subject(s)
Blood Pressure , Circadian Rhythm , Heart Rate , Photoperiod , Analysis of Variance , Animals , Blood Pressure Monitoring, Ambulatory , Circadian Rhythm/physiology , Heart Rate Determination , Linear Models , Male , Nonlinear Dynamics , Photic Stimulation , Rats, Wistar , Telemetry
17.
Chronobiol Int ; 33(9): 1148-1160, 2016.
Article in English | MEDLINE | ID: mdl-27459109

ABSTRACT

Disturbances in regular circadian oscillations can have negative effects on cardiovascular function, but epidemiological data are inconclusive and new data from animal experiments elucidating critical biological mechanisms are needed. To evaluate the consequences of chronic phase shifts of the light/dark (LD) cycle on hormonal and cardiovascular rhythms, two experiments were performed. In Experiment 1, male rats were exposed to either a regular 12:12 LD cycle (CONT) or rotating 8-h phase-delay shifts of LD every second day (SHIFT) for 10 weeks. During this period, blood pressure (BP) was monitored weekly, and daily rhythms of melatonin, corticosterone, leptin and testosterone were evaluated at the end of the experiment. In Experiment 2, female rats were exposed to the identical shifted LD schedule for 12 weeks, and daily rhythms of BP, heart rate (HR) and locomotor activity were recorded using telemetry. Preserved melatonin rhythms were found in the pineal gland, plasma, heart and kidney of SHIFT rats with damped amplitude in the plasma and heart, suggesting that the central oscillator can adapt to chronic phase-delay shifts. In contrast, daily rhythms of corticosterone, testosterone and leptin were eliminated in SHIFT rats. Exposure to phase shifts did not lead to increased body weight and elevated BP. However, a shifted LD schedule substantially decreased the amplitude and suppressed the circadian power of the daily rhythms of BP and HR, implying weakened circadian control of physiological and behavioural processes. The results demonstrate that endocrine and cardiovascular rhythms can differentially adapt to chronic phase-delay shifts, promoting internal desynchronization between central and peripheral oscillators, which in combination with other negative environmental stimuli may result in negative health effects.


Subject(s)
Blood Pressure , Circadian Rhythm , Endocrine Glands/physiology , Heart Rate , Light , Locomotion/physiology , Animals , Corticosterone/metabolism , Female , Leptin/metabolism , Male , Melatonin/metabolism , Random Allocation , Rats , Rats, Wistar , Testosterone/metabolism
18.
Hypertens Res ; 39(7): 501-5, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26911229

ABSTRACT

Decreased oxygenation during pregnancy and early periods of ontogeny can affect normal body development and result in diseases in adulthood. The aim of this study was to use the model of prenatal intermittent hypoxia (PIH) and evaluate the effects of short-term hypoxia at the end of gestation on blood pressure (BP) control in adulthood. Wistar rats were exposed daily to PIH for 4 h during gestational day 19 and 20. In adult male rats, heart rate (HR), systolic BP and pulse pressure (PP) were acquired by radiotelemetry during 1 week. On the basis of HR variability and BP variability, sympathovagal balance (LF/HF) and spontaneous baroreflex sensitivity (sBRS) were evaluated. Systolic BP and PP were significantly elevated in PIH rats in comparison with control rats during the light and dark phase of the day, while LF/HF increased only during the light phase of the day. In contrast, sBRS tended to decrease only during the dark phase in PIH rats. In all measured and calculated parameters, significant circadian rhythms were present and were not affected by PIH. In conclusion, our data suggest that short intermittent hypoxia at the end of gestation can increase BP and PP via significant changes in LF/HF, which occur especially during the passive phase of the day. Results suggest that minor changes in the autonomous nervous system activity induced by environmental conditions during the perinatal period may contribute to development of hypertension in adulthood.


Subject(s)
Blood Pressure , Circadian Rhythm , Fetal Hypoxia/complications , Heart Rate , Hypertension/etiology , Animals , Autonomic Nervous System/physiopathology , Blood Pressure/physiology , Circadian Rhythm/physiology , Female , Fetal Hypoxia/physiopathology , Heart Rate/physiology , Hypertension/physiopathology , Male , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar
19.
Physiol Behav ; 156: 16-23, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26773465

ABSTRACT

Disturbed light-dark (LD) cycles are associated with circadian disruption of physiological and behavioural rhythms and in turn with an increased risk of disease development. However, direct causal links and underlying mechanisms leading to negative health consequences still need to be revealed. In the present study, we exposed male Wistar rats to repeated phase shifts of LD cycle and analysed their ability to cope with mild emotional stressors. In experiment 1, rats were submitted to either a regular 12:12 LD cycle (CTRL rats) or 8-h phase delay shifts applied every 2days for 5weeks (SHIFT rats). Subsequently, the behaviour was examined in the open-field, black-white box and elevated plus maze tests. In experiment 2, changes in blood pressure (BP), heart rate (HR) as well as the activity of autonomic nervous system were measured in telemeterised rats in response to open-field and black-white box tests before and after 5-week exposure to shifted LD regime. Locomotor activity was consistently higher in SHIFT than CTRL rats in in the open-field and black-white box tests. Interestingly, in the elevated plus maze, SHIFT rats displayed increased risk assessment and decreased grooming compared to CTRL rats. Anxiety measures were affected only in the black-white box, where SHIFT rats displayed reduced anxiety-like behaviour compared to CTRL rats. Differences in behavioural reactivity between SHIFT and CTRL rats did not correspond with BP and HR changes. However, exposure to phase shifts increased the sympathovagal reactivity in the black-white box. Together, our results demonstrated that disturbed LD conditions decreased emotional reactivity of rats and affected their ability to cope with emotional stressors denoting an additional risk mechanism linking disrupted circadian organisation to adverse health effects.


Subject(s)
Circadian Rhythm/physiology , Emotions/physiology , Photoperiod , Animals , Autonomic Nervous System , Blood Pressure/physiology , Heart Rate/physiology , Male , Motor Activity/physiology , Rats , Rats, Wistar/physiology , Rats, Wistar/psychology
20.
Clin Exp Hypertens ; 37(2): 142-7, 2015.
Article in English | MEDLINE | ID: mdl-25050593

ABSTRACT

A direct relationship exists between salt consumption and hypertension. Increased sodium intake does not automatically lead to a rise in blood pressure (BP) because of marked intra-individual variability in salt sensitivity. Wistar rats are a salt-resistant strain and increased salt intake in adults does not induce hypertension. Mechanisms regulating BP develop during early ontogenesis and increased sodium consumption by pregnant females leads to an increase in BP of their offspring, but early postnatal stages have not been sufficiently analyzed in salt-resistant strains of rats. The aim of this work was to study the effects of increased salt during early ontogeny on cardiovascular characteristics of Wistar rats. We used 16 control (C; 8 males + 8 females) rats fed with a standard diet (0.2% sodium) and 16 experimental (S; 8 males + 8 females) rats fed with a diet containing 0.8% sodium. BP was measured weekly and plasma renin activity, aldosterone and testosterone concentrations were assayed by radioimmunoassay after the experiment in 16-week-old animals. In the kidney, AT1 receptors were determined by the western blot. BP was higher in the S as compared with the C rats and did not differ between males and females. The relative left ventricle mass was increased in S as compared with C males and no differences were recorded in females. No significant differences between groups were found in hormonal parameters and AT1 receptors. Results indicate that moderately increased salt intake during postnatal ontogeny results in a BP rise even in salt-resistant rats.


Subject(s)
Blood Pressure/drug effects , Hypertension/chemically induced , Pregnancy, Animal , Sodium Chloride, Dietary/toxicity , Sodium/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Aldosterone/blood , Animals , Blotting, Western , Disease Models, Animal , Female , Hypertension/metabolism , Hypertension/physiopathology , Male , Pregnancy , Radioimmunoassay , Rats , Rats, Wistar , Renin/blood , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...