Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Can J Physiol Pharmacol ; 100(4): 324-333, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34670103

ABSTRACT

Functional and structural adaptation of common carotid artery could be one of the important causes of postflight orthostatic intolerance after microgravity exposure, the mechanisms of which remain unclear. Recent evidence indicates that long-term spaceflight increases carotid artery stiffness, which might present a high risk to astronaut health and postflight working ability. Studies have suggested that vascular calcification is a common pathological change in cardiovascular diseases that is mainly manifested as an increase in vascular stiffness. Therefore, this study investigated whether simulated microgravity induces calcification of common carotid artery and to elucidate the underlying mechanisms. Four-week-old hindlimb-unweighted (HU) rats were used to simulate the deconditioning effects of microgravity on cardiovascular system. We found that simulated microgravity induced vascular smooth muscle cell (VSMC) osteogenic differentiation and medial calcification, increased receptor activator of nuclear factor κB (NF-κB) ligand (RANKL) and RANK expression, and enhanced NF-κB activation in rat common carotid artery. In vitro activation of the RANK pathway with exogenous RANKL, a RANK ligand, increased RANK and osteoprotegerin (OPG) expression in HU rats. Moreover, the expression of osteogenic markers and activation of NF-κB in HU rats were further enhanced by exogenous RANKL but suppressed by the RANK inhibitor osteoprotegerin fusion protein (OPG-Fc). These results indicated that the OPG/RANKL/RANK system modulates VSMC osteogenic differentiation and medial calcification of common carotid artery in simulated microgravity rats by regulating the NF-kB pathway.


Subject(s)
Osteoprotegerin , Weightlessness , Animals , Carotid Artery, Common/metabolism , NF-kappa B/metabolism , Osteogenesis , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Rats , Weightlessness/adverse effects
2.
Cell Prolif ; 53(3): e12774, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32034930

ABSTRACT

OBJECTIVES: Postflight orthostatic intolerance has been regarded as a major adverse effect after microgravity exposure, in which cerebrovascular adaptation plays a critical role. Our previous finding suggested that dedifferentiation of vascular smooth muscle cells (VSMCs) might be one of the key contributors to cerebrovascular adaptation under simulated microgravity. This study was aimed to confirm this concept and elucidate the underlying mechanisms. MATERIALS AND METHODS: Sprague Dawley rats were subjected to 28-day hindlimb-unloading to simulate microgravity exposure. VSMC dedifferentiation was evaluated by ultrastructural analysis and contractile/synthetic maker detection. The role of T-type CaV 3.1 channel was revealed by assessing its blocking effects. MiR-137 was identified as the upstream of CaV 3.1 channel by luciferase assay and investigated by gain/loss-of-function approaches. Calcineurin/nuclear factor of activated T lymphocytes (NFAT) pathway, the downstream of CaV 3.1 channel, was investigated by detecting calcineurin activity and NFAT nuclear translocation. RESULTS: Simulated microgravity induced the dedifferentiation and proliferation in rat cerebral VSMCs. T-type CaV 3.1 channel promoted the dedifferentiation and proliferation of VSMC. MiR-137 and calcineurin/NFATc3 pathway were the upstream and downstream signalling of T-type CaV 3.1 channel in modulating the dedifferentiation and proliferation of VSMCs, respectively. CONCLUSIONS: The present work demonstrated that miR-137 and its target T-type CaV 3.1 channel modulate the dedifferentiation and proliferation of rat cerebral VSMCs under simulated microgravity by regulating calcineurin/NFATc3 pathway.


Subject(s)
Calcineurin/metabolism , Calcium Channels, T-Type/metabolism , Cerebral Arteries/cytology , MicroRNAs/metabolism , Myocytes, Smooth Muscle/cytology , NFATC Transcription Factors/metabolism , Animals , Brain/blood supply , Calcium Channels, T-Type/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cerebral Arteries/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Weightlessness Simulation
3.
Int J Mol Sci ; 20(16)2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31416128

ABSTRACT

The functional and structural adaptations in cerebral arteries could be one of the fundamental causes in the occurrence of orthostatic intolerance after space flight. In addition, emerging studies have found that many cardiovascular functions exhibit circadian rhythm. Several lines of evidence suggest that space flight might increase an astronaut's cardiovascular risks by disrupting circadian rhythm. However, it remains unknown whether microgravity disrupts the diurnal variation in vascular contractility and whether microgravity impacts on circadian clock system. Sprague-Dawley rats were subjected to 28-day hindlimb-unweighting to simulate the effects of microgravity on vasculature. Cerebrovascular contractility was estimated by investigating vasoconstrictor responsiveness and myogenic tone. The circadian regulation of CaV1.2 channel was determined by recording whole-cell currents, evaluating protein and mRNA expressions. Then the candidate miRNA in relation with Ca2+ signal was screened. Lastly, the underlying pathway involved in circadian regulation of cerebrovascular contractility was determined. The major findings of this study are: (1) The clock gene BMAL1 could induce the expression of miR-103, and in turn modulate the circadian regulation of CaV1.2 channel in rat cerebral arteries at post-transcriptional level; and (2) simulated microgravity disrupted intrinsic diurnal oscillation in rat cerebrovascular contractility by altering circadian regulation of BMAL1/miR-103/CaV1.2 signal pathway.


Subject(s)
ARNTL Transcription Factors/genetics , Calcium Channels, L-Type/metabolism , Cerebrovascular Circulation/genetics , Circadian Rhythm , MicroRNAs/genetics , Vasoconstriction/genetics , Weightlessness , ARNTL Transcription Factors/metabolism , Animals , Cell Line , Gene Expression Regulation , Male , Models, Biological , Rats , Signal Transduction
4.
Can J Physiol Pharmacol ; 96(8): 772-782, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29527943

ABSTRACT

Recent studies have suggested that microgravity-induced arterial remodelling contributes to post-flight orthostatic intolerance and that multiple mechanisms are involved in arterial remodelling. However, the initial mechanism by which haemodynamic changes induce arterial remodelling is unknown. Focal adhesions (FAs) are dynamic protein complexes that have mechanotransduction properties. This study aimed to investigate the role of FAs in simulated-microgravity-induced basilar and femoral arterial remodelling. A 4-week hindlimb-unweighted (HU) rat model was used to simulate the effects of microgravity, and daily 1-hour intermittent artificial gravity (IAG) was used to prevent arterial remodelling. After 4-week HU, wall thickness, volume of smooth muscle cells (SMCs) and collagen content were increased in basilar artery but decreased in femoral artery (P < 0.05). Additionally, the expression of p-FAK Y397 and p-Src Y418 was increased and reduced in SMCs of basilar and femoral arteries, respectively, by HU (P < 0.05). The number of FAs was increased in basilar artery and reduced in femoral artery by HU (P < 0.05). Furthermore, daily 1-hour IAG prevented HU-induced differential structural adaptations and changes in FAs of basilar and femoral arteries. These results suggest that FAs may act as mechanosensors in arterial remodelling by initiating intracellular signal transduction in response to altered mechanical stress induced by microgravity.


Subject(s)
Basilar Artery/physiology , Femoral Artery/physiology , Focal Adhesions/metabolism , Vascular Remodeling , Weightlessness Simulation , Adaptation, Physiological , Animals , Cerebral Arteries/physiology , Collagen/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hindlimb Suspension , Male , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Rats, Sprague-Dawley , src-Family Kinases/metabolism
5.
PLoS One ; 9(5): e97737, 2014.
Article in English | MEDLINE | ID: mdl-24840155

ABSTRACT

BACKGROUND: To elucidate further from the biomechanical aspect whether microgravity-induced cerebral vascular mal-adaptation might be a contributing factor to postflight orthostatic intolerance and the underlying mechanism accounting for the potential effectiveness of intermittent artificial gravity (IAG) in preventing this adverse effect. METHODOLOGY/PRINCIPAL FINDINGS: Middle cerebral arteries (MCAs) were isolated from 28-day SUS (tail-suspended, head-down tilt rats to simulate microgravity effect), S+D (SUS plus 1-h/d -Gx gravitation by normal standing to simulate IAG), and CON (control) rats. Vascular myogenic reactivity and circumferential stress-strain and axial force-pressure relationships and overall stiffness were examined using pressure arteriography and calculated. Acellular matrix components were quantified by electron microscopy. The results demonstrate that myogenic reactivity is susceptible to previous pressure-induced, serial constrictions. During the first-run of pressure increments, active MCAs from SUS rats can strongly stiffen their wall and maintain the vessels at very low strains, which can be prevented by the simulated IAG countermeasure. The strains are 0.03 and 0.14 respectively for SUS and S+D, while circumferential stress being kept at 0.5 (106 dyn/cm2). During the second-run pressure steps, both the myogenic reactivity and active stiffness of the three groups declined. The distensibility of passive MCAs from S+D is significantly higher than CON and SUS, which may help to attenuate the vasodilatation impairment at low levels of pressure. Collagen and elastin percentages were increased and decreased, respectively, in MCAs from SUS and S+D as compared with CON; however, elastin was higher in S+D than SUS rats. CONCLUSIONS: Susceptibility to previous myogenic constrictions seems to be a self-limiting protective mechanism in cerebral small resistance arteries to prevent undue cerebral vasoconstriction during orthostasis at 1-G environment. Alleviating of active stiffening and increasing of distensibility of cerebral resistance arteries may underlie the countermeasure effectiveness of IAG.


Subject(s)
Middle Cerebral Artery/anatomy & histology , Middle Cerebral Artery/physiology , Weightlessness Simulation/methods , Angiography , Animals , Biomechanical Phenomena , Microscopy, Electron , Pressure , Rats
6.
Sheng Li Xue Bao ; 64(2): 107-20, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22513459

ABSTRACT

The aim of the present study was to evaluate the active and passive mechanical properties and wall collagen and elastin contents of mesenteric small arteries (MSAs) isolated from rats of 28-day simulated microgravity (SUS), countermeasure [S + D: SUS plus 1 h/d -G(x) to simulate intermittent artificial gravity (IAG)] and control (CON) groups. Three mechanical parameters were calculated: the overall stiffness (ß), circumferential stress (σ(θ))-strain (ε(θ)) relationship and pressure-dependent incremental elastic modulus (E(inc,p)). Vessel wall collagen and elastin percentage were quantified by electron microscopy. The results demonstrate that the active mechanical behavior of MSAs differs noticeably among the three groups: the active stress-strain curve of SUS vessels is very close to the passive curve, whereas the active σ(θ)-ε(θ) curves of CON and S + D vessels are shifted leftward and display a parabolic shape, indicating that for MSAs isolated from S + D, but not those from SUS rats, the pressure-induced myogenic constriction can effectively stiffen the vessel wall as the CON vessels. The passive mechanical behavior of MSAs does not show significant differences among the three groups. However, the percentage of collagen is decreased in the wall of SUS and S + D compared with CON vessels in the following order: SUS < S + D < CON. Thus, the relationship between passive mechanical behavior and compositional changes may be complex and yet depends on factors other than the quantity of collagen and elastin. These findings have provided biomechanical data for the understanding of the mechanism of postflight orthostatic intolerance and its gravity-based countermeasure.


Subject(s)
Mesenteric Arteries/physiology , Vasoconstriction/physiology , Weightlessness Simulation , Animals , Biomechanical Phenomena , Collagen/metabolism , Elasticity , Hindlimb Suspension/physiology , Male , Mesenteric Arteries/ultrastructure , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/ultrastructure , Random Allocation , Rats , Rats, Sprague-Dawley , Stress, Mechanical
7.
Sheng Li Xue Bao ; 64(1): 14-26, 2012 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-22348956

ABSTRACT

The present study was designed to test the hypothesis that a medium-term simulated microgravity can induce region-specific remodeling in large elastic arteries with their innermost smooth muscle (SM) layers being most profoundly affected. The second purpose was to examine whether these changes can be prevented by a simulated intermittent artificial gravity (IAG). The third purpose was to elucidate whether vascular local renin-angiotensin system (L-RAS) plays an important role in the regional vascular remodeling and its prevention by the gravity-based countermeasure. This study consisted of two interconnected series of in-vivo and ex-vivo experiments. In the in-vivo experiments, the tail-suspended, hindlimb unloaded rat model was used to simulate microgravity-induced cardiovascular deconditioning for 28 days (SUS group); and during the simulation period, another group was subjected to daily 1-hour dorso-ventral (-G(x)) gravitation provided by restoring to normal standing posture (S + D group). The activity of vascular L-RAS was evaluated by examining the gene and protein expression of angiotensinogen (Ao) and angiotensin II receptor type 1 (AT1R) in the arterial wall tissue. The results showed that SUS induced an increase in the media thickness of the common carotid artery due to hypertrophy of the four SM layers and a decrease in the total cross-sectional area of the nine SM layers of the abdominal aorta without significant change in its media thickness. And for both arteries, the most prominent changes were in the innermost SM layers. Immunohistochemistry and in situ hybridization revealed that SUS induced an up- and down-regulation of Ao and AT1R expression in the vessel wall of common carotid artery and abdominal aorta, respectively, which was further confirmed by Western blot analysis and real time PCR analysis. Daily 1-hour restoring to normal standing posture over 28 days fully prevented these remodeling and L-RAS changes in the large elastic arteries that might occur due to SUS alone. In the ex-vivo experiments, to elucidate the important role of transmural pressure in vascular regional remodeling and differential regulation of L-RAS activity, we established an organ culture system in which rat common carotid artery, held at in-vivo length, can be perfused and pressurized at varied flow and pressure for 7 days. In arteries perfused at a flow rate of 7.9 mL/min and pressurized at 150 mmHg, but not at 0 or 80 mmHg, for 3 days led to an augmentation of c-fibronectin (c-FN) expression, which was also more markedly expressed in the innermost SM layers, and an increase in Ang II production detected in the perfusion fluid. However, the enhanced c-FN expression and increased Ang II production that might occur due to a sustained high perfusion pressure alone were fully prevented by daily restoration to 0 or 80 mmHg for a short duration. These findings from in-vivo and ex-vivo experiments have provided evidence supporting our hypothesis that redistribution of transmural pressures might be the primary factor that initiates region-specific remodeling of arteries during microgravity and the mechanism of IAG is associated with an intermittent restoration of the transmural pressures to their normal distribution. And they also provide support to the hypothesis that L-RAS plays an important role in vascular adaptation to microgravity and its prevention by the IAG countermeasure.


Subject(s)
Angiotensinogen/metabolism , Aorta, Abdominal/pathology , Carotid Artery, Common/pathology , Receptor, Angiotensin, Type 1/metabolism , Weightlessness Simulation , Angiotensinogen/genetics , Animals , Aorta, Abdominal/physiopathology , Carotid Artery, Common/physiopathology , Hindlimb Suspension , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System/physiology
8.
Sheng Li Xue Bao ; 63(4): 311-8, 2011 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-21861049

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder characterized clinically by rigidity, akinesia, resting tremor and postural instability. It has recently been suggested that low frequency stimulation of the pedunculopontine nucleus (PPN) has a role in the therapy for Parkinsonism, particularly in gait disorder and postural instability. However, there is limited information about the mechanism of low frequency stimulation of the PPN on Parkinson's disease. The present study was to investigate the effect and mechanism of low frequency stimulation of the PPN on the firing rate of the ventrolateral thalamic nucleus (VL) in a rat model with unilateral 6-hydroxydopamine lesioning of the substantia nigra pars compacta. In vivo extracellular recording and microiontophoresis were adopted. The results showed that the firing rate of 60.71% VL neurons in normal rats and 59.57% VL neurons in 6-hydroxydopamine lesioned rats increased with low frequency stimulation of the PPN. Using microiontophoresis to VL neurons, we found the firing rate in VL neurons responded with either an increase or decrease in application of acetylcholine (ACh) in normal rats, whereas with a predominant decrease in M receptor antagonist atropine. Furthermore, the VL neurons were mainly inhibited by application of γ-aminobutyric acid (GABA) and excited by GABA(A) receptor antagonist bicuculline. Importantly, the VL neurons responding to ACh were also inhibited by application of GABA. We also found that the excitatory response of the VL neurons to the low frequency stimulation of the PPN was significantly reversed by microiontophoresis of atropine. These results demonstrate that cholinergic and GABAergic afferent nerve fibers may converge on the same VL neurons and they are involved in the effects of low frequency stimulation of the PPN, with ACh combining M(2) receptors on the presynaptic membrane of GABAergic afferents, which will inhibit the release of GABA in the VL and then improve the symptoms of Parkinson's disease.


Subject(s)
Action Potentials , Electric Stimulation , Parkinson Disease, Secondary/physiopathology , Pedunculopontine Tegmental Nucleus/physiology , Ventral Thalamic Nuclei/physiology , Acetylcholine/metabolism , Animals , Cholinergic Fibers/physiology , Male , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/therapy , Rats , Rats, Sprague-Dawley
9.
Am J Physiol Cell Physiol ; 298(6): C1489-500, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20457834

ABSTRACT

Cerebral arterial remodeling is one of the critical factors in the occurrence of postspaceflight orthostatic intolerance. We hypothesize that large-conductance calcium-activated K(+) (BK(Ca)) channels in vascular smooth muscle cells (VSMCs) may play an important role in regulating cerebrovascular adaptation during microgravity exposure. The aim of this work was to investigate whether activation of BK(Ca) channels is involved in regulation of apoptotic remodeling of cerebral arteries in simulated microgravity rats. In animal studies, Sprague-Dawley rats were subjected to 1-wk hindlimb unweighting to simulate microgravity. Alterations of BK(Ca) channels in cerebral VSMCs were investigated by patch clamp and Western blotting; apoptosis was assessed by electron microscopy and terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick-end labeling (TUNEL). To evaluate the correlation of BK(Ca) channel and apoptosis, channel protein and cell nucleus were double-stained. In cell studies, hSloalpha+beta1 channel was coexpressed into human embryonic kidney 293 (HEK293) cells to observe the effects of BK(Ca) channels on apoptosis. In rats, enhanced activities and expression of BK(Ca) channels were found to be correlated with increased apoptosis in cerebral VSMCs after simulated microgravity. In transfected HEK293 cells, activation of cloned BK(Ca) channel induced apoptosis, whereas inhibition of cloned BK(Ca) channel decreased apoptosis. In conclusion, activation of BK(Ca) channels is associated with increased apoptosis in cerebral VSMCs of simulated microgravity rats.


Subject(s)
Apoptosis , Calcium/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Weightlessness Simulation , Animals , Blotting, Western , Cell Line , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Hindlimb Suspension , Humans , In Situ Nick-End Labeling , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Potentials , Microscopy, Electron , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Transfection
10.
Apoptosis ; 15(4): 426-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20012488

ABSTRACT

The large conductance Ca(2+)-activated K(+) (BK(Ca)) channels are highly expressed in vascular smooth muscle cells (VSMCs) and play an essential role in the regulation of various physiological functions. Besides its electrophysiological function in vascular relaxation, BK(Ca) has also been reported to be implicated in nitric oxide (NO)-induced apoptosis of VSMCs. However, the molecular mechanism is not clear and has not been determined on cloned channels. The present study was designed to clarify whether activation of cloned BK(Ca) channel was involved in NO-induced apoptosis in human embryonic kidney 293 (HEK293) cell. The cDNA encoding the alpha-subunit of BK(Ca) channel, hSloalpha, was transiently transfected into HEK293 cells. The apoptotic death in HEK-hSloalpha cells was detected using immunocytochemistry, analysis of fragmented DNA by agarose gel electrophoresis, MTT test, and flow cytometry assays. Whole-cell and single-channel characteristics of HEK-hSloalpha cells exhibited functional features similar to native BK(Ca) channel in VSMCs. Exposuring of HEK- hSloalpha cells to S-nitroso-N-acetyl-penicillamine increased the hSloalpha channel activities of whole-cell and single-channel, and then increased percentage of cells undergoing apoptosis. However, blocking hSloalpha channels with 1 mM tetraethylammonia or 100 nM iberiotoxin significantly decreased the NO-induced apoptosis, whereas 30 microM NS1619, the specific agonist of BK(Ca), independently increased hSloalpha currents and induced apoptosis. These results indicated that activation of cloned BK(Ca) channel was involved in NO-induced apoptosis of HEK293 cells.


Subject(s)
Apoptosis/drug effects , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Nitric Oxide/pharmacology , Cell Line , Cloning, Molecular , Humans , Patch-Clamp Techniques , Peptides/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Tetraethylammonium/pharmacology , Transfection
11.
Electrophoresis ; 30(15): 2600-12, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19670232

ABSTRACT

In this study, novel polysulfone-based cation-exchange membranes with strong mechanical strength have been developed and applied in ion-exchange-membrane-partitioned free-flow IEF (IEM-FFIEF) to replace the conventional immobiline membranes. A fundamental understanding of protein mass transfer in the IEM-FFIEF process has been revealed experimentally with the aid of membrane-based boundary effect model contributed by Ennis et al. we have proven experimentally the existence of a pH gradient across the membrane cross-section when an IEM-FFIEF system is in operation. The boundary effects on particle velocities are calculated based on the IEF assumption and various characterizations, and are compared with the experimental results. In the IEM-FFIEF experiments, a protein mixture (BSA and myoglobin (Mb)) and sulfonated polysulfone membranes with different ion-exchange capacities are applied. Experimental results show that the real velocity and real mobility (of Mb in this study) are comparable with the mathematic model developed by Ennis et al. This suggests that the equation proposed by Ennis et al., is sufficient to capture the mass transfer through membrane in the IEM-FFIEF system after considering the effects of pore size distribution and effects of disturbed electric field. The charge properties of the membrane surface play a dominant role on the separation performance of the membranes. The newly developed porous solid-phase ion-exchange membranes may potentially and effectively replace immobilines to perform the selective function for protein separation.


Subject(s)
Chromatography, High Pressure Liquid/methods , Immunoelectrophoresis/methods , Membranes, Artificial , Proteins/isolation & purification , Chromatography, Ion Exchange , Electromagnetic Phenomena , Hydrogen-Ion Concentration , Models, Chemical , Polymers/chemistry , Porosity , Proteins/chemistry , Proton-Motive Force , Sulfones/chemistry
12.
Sheng Li Xue Bao ; 61(4): 386-94, 2009 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-19701592

ABSTRACT

The aim of the present study was to further elucidate the mechanisms of vascular adaptation to microgravity and its gravity-based countermeasure by a biomechanical approach. Active (the dissected vessel segment was superfused with PPS) and passive (while it was superfused with Ca(2+)-free PPS) biomechanical properties of mesenteric third-order small arteries and middle cerebral arteries isolated from 3-day simulated microgravity (SUS), countermeasure (STD, daily 1 h of -G(x) gravitation), and control (CON) groups of rats were studied. The following mechanical parameters were calculated: the overall stiffness parameter of passive vessels (beta), circumferential stress (sigma(theta))-strain (epsilon(theta)) relationship, and pressure-dependent incremental elastic modulus (E(inc,p)) of both active and passive vessels, and vascular smooth muscle (VSM) activity-dependent incremental modulus (E(inc,a)). Results from the analysis of active biomechanical properties revealed the contribution of vascular smooth muscle (VSM) tone during the early adaptation to microgravity: (1) For mesenteric small arteries, active circumferential sigma(theta) -epsilon(theta) curve of SUS group was comparable with that of the passive vessels, indicating that the function of VSM to restore the normal stress distribution is compromised; however, this mal-adaptation was fully prevented by the countermeasure of daily 1 h of -G(x) gravitation; (2) For the middle cerebral arteries, active circumferential sigma(theta) -epsilon(theta) relation of SUS group was shifted to the left side of the passive curve and epsilon(theta) was kept at a nearly constant level with the corresponding sigma(theta) being at its normal range; furthermore, the enhanced myogenic tone responsiveness was not prevented by daily short-duration -G(x). Analysis of the passive biomechanical properties has suggested remodeling changes in matrix components of different types of vessels, which might be significant if the exposure duration was further prolonged. In brief, studies of vascular biomechanics are of particular importance in elucidating the mechanisms underlying vascular adaptation to microgravity and its gravity-based countermeasure.


Subject(s)
Mesenteric Arteries/physiology , Middle Cerebral Artery/physiology , Weightlessness Simulation , Animals , Biomechanical Phenomena , Muscle, Smooth, Vascular/physiology , Pressure , Rats
13.
Mol Cell Biochem ; 331(1-2): 117-26, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19430934

ABSTRACT

Large-conductance Ca(2+)-activated K(+) channel is formed by a tetramer of the pore-forming alpha-subunit and distinct accessory beta-subunits (beta1-beta4) which contribute to BK(Ca) channel molecular diversity. Accumulative evidences indicate that not only alpha-subunit alone but also the alpha + beta subunit complex and/or beta-subunit might play an important role in modulating various physiological functions in most mammalian cells. To evaluate the detailed pharmacological and biophysical properties of alpha + beta1 subunit complex or beta1-subunit in BK(Ca) channel, we established an expression system that reliably coexpress hSloalpha + beta1 subunit complex in HEK293 cells. The coexpression of hSloalpha + beta1 subunit complex was evaluated by western blotting and immunolocalization, and then the single-channel kinetics and pharmacological properties of expressed hSloalpha + beta1 subunit complex were investigated by cell-attached and outside-out patches, respectively. The results in this study showed that the expressed hSloalpha + beta1 subunit complex demonstrated to be fully functional for its typical single-channel traces, Ca(2+)-sensitivity, voltage-dependency, high conductance (151 +/- 7 pS), and its pharmacological activation and inhibition.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Benzimidazoles/pharmacology , Blotting, Western , Calcium/pharmacology , Cell Line , Cell Shape/drug effects , Humans , Ion Channel Gating/drug effects , Patch-Clamp Techniques , Peptides/pharmacology , Plasmids/genetics , Transfection
14.
J Appl Physiol (1985) ; 106(1): 251-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18988766

ABSTRACT

Previous studies have demonstrated activation of the local renin-angiotensin system in hindlimb unweighting (HU) rat vasculature. The present study intended to identify the effects of blockade of angiotensin II (ANG II) type 1 (AT(1)) receptors with losartan on vascular reactivity, nitric oxide synthase (NOS) expression, and superoxide anion (O(2)(*-)) levels in 3-wk HU rat cerebral and carotid arteries. Three weeks later, vasoconstriction, vasodilatation, endothelial NOS (eNOS) and inducible NOS (iNOS) protein, as well as O(2)(*-) levels in rat cerebral and carotid arteries were examined. We found that HU enhanced maximal response to KCl/5-hydroxytryptamine (P < 0.01) in basilar arteries and KCl/phenylephrine (P < 0.05) in common carotid arteries from HU rats. Acetylcholine induced concentration-dependent vasodilatation in all the artery rings, but with significantly smaller amplitude in basilar (P < 0.01) and common carotid (P < 0.05) arteries from HU rats than those from control rats. Chronic treatment with losartan partially restored response to vasoconstrictors and acetylcholine-induced vasodilatation in basilar (P < 0.01) and common carotid (P < 0.05) arteries from losartan-treated HU rats. Furthermore, iNOS content in cerebral arteries and eNOS/iNOS content in carotid arteries were significantly (P < 0.01) increased in HU rats. Meanwhile, HU increased O(2)(*-) levels in all the layers of these arteries. However, losartan restored NOS content and O(2)(*-) levels toward normal. These results suggested that the HU-induced enhancement of vasoconstriction and reduction in endothelium-dependent relaxation involved alterations in O(2)(*-) and NOS content through an ANG II/AT(1) receptor signaling pathway.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Basilar Artery/drug effects , Carotid Artery, Common/drug effects , Hindlimb Suspension , Losartan/pharmacology , Nitric Oxide Synthase/metabolism , Superoxides/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects , Angiotensin II/metabolism , Animals , Basilar Artery/metabolism , Carotid Artery, Common/metabolism , Dose-Response Relationship, Drug , Male , Models, Animal , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Weightlessness Simulation
15.
J Appl Physiol (1985) ; 105(4): 1134-45, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18703761

ABSTRACT

This study was designed to test the hypothesis that postsuspension cardiovascular manifestation in conscious rats after a medium-term (28-day) tail suspension (SUS) is hypertensive and tachycardiac and can be prevented by a countermeasure of daily 1-h dorsoventral (-G(x)) gravitation provided by standing (STD). To assess associated changes in cardiovascular regulation, blood pressure (BP) and heart rate (HR) variability were analyzed by spectral analysis computed by parametric autoregressive (AR) method and by nonlinear recurrence quantification analysis (RQA) and approximate entropy (ApEn) measure. The results showed that conscious SUS rats manifested hypertensive and tachycardiac response before and after being released from suspension compared with the controls, and the countermeasure of 1 h/day -G(x) prevented the hypertensive response. Auto- and cross-spectral analysis and transfer function analysis did not show significant changes in cardiovascular variability. However, SUS decreased the three RQA indexes [recurrence percentage (RC%), determinism percentage (DT%), and the longest diagonal line (L(max))] of systolic BP, whereas STD alleviated these changes. ApEn values of HR data sets were significantly higher in the SUS and SUS + STD groups compared with those of the control group before and after release from suspension. The present study has demonstrated that daily -G(x) for as short as 1 h is sufficient to prevent postsuspension cardiovascular alteration in conscious rats after a medium-term SUS. Nonlinear measures, but not spectral analysis, might provide promising data to estimate the overall changes in cardiovascular autonomic regulation due to microgravity exposure.


Subject(s)
Autonomic Nervous System/physiopathology , Blood Pressure , Cardiovascular System/innervation , Heart Rate , Hypertension/prevention & control , Tachycardia/prevention & control , Weightlessness Countermeasures , Weightlessness Simulation/adverse effects , Animals , Hindlimb Suspension/adverse effects , Hypertension/etiology , Hypertension/physiopathology , Male , Models, Animal , Models, Cardiovascular , Nonlinear Dynamics , Rats , Rats, Sprague-Dawley , Signal Processing, Computer-Assisted , Tachycardia/etiology , Tachycardia/physiopathology , Time Factors
16.
Sheng Li Xue Bao ; 60(1): 74-82, 2008 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-18288361

ABSTRACT

The aim of the present study was to examine whether there are changes in systolic and diastolic blood pressure (SBP and DBP) and heart rate (HR) and their spectral indices in conscious free-moving rats after tail-suspension for 28 d. The tail-suspended hindlimb-unloaded (HU) rat model was used to simulate the cardiovascular effect of microgravity and the post-spaceflight cardiovascular dysfunction. The auto- and cross-spectral analysis of SBP variability (SBPV) and HR variability (HRV) were performed by the method based on the autoregressive model (AR), and the auto-spectral results was compared with the results from the classical periodogram method. The baroreceptor-heart rate reflex sensitivity (BRS) was estimated using transfer function analysis from SBP to HR. The results indicated that auto-spectral results based on the two methods were comparable, while smoother power spectral curves with distinguished peaks were trained by the AR method. The means of SBP, DBP, and HR, the main spectral indices of SBPV and HRV, and the mean average gain of transfer functions computed at low- and high-frequency ranges (0.25-0.8 Hz and 0.8-2.4 Hz) did not show significant changes before and after release from suspension. Furthermore, the main spectral indices of SBPV and HRV at different time points did not show significant differences between the control and suspension groups. However, the means of SBP, DBP, and HR at different time points were significantly higher in simulated weightless rats than those in the control rats. The findings of the present study suggest that a mid-term simulated microgravity might induce hypertension and tachycardia upon removal from the suspension which reflects a general sympathetic hyperactivity. We speculated that the sympathetic hyperactivity might be a compensatory mechanism activated in the intact animal to counteract HU-induced hypo-responsiveness of resistance vessels. In addition, lack of clear and distinct changes in HRV and BRS have also been reported in some recent space and ground-based human studies.


Subject(s)
Blood Pressure , Heart Rate , Weightlessness Simulation , Animals , Baroreflex , Hindlimb Suspension , Hypertension , Rats
17.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 22(3): 269-73, 2006 Aug.
Article in Chinese | MEDLINE | ID: mdl-21158066

ABSTRACT

AIM: To investigate the effects of simulated microgravity on dilatory responsiveness and NOS expression of abdominal aorta in rats. METHODS: Twenty male healthy SD rats, which body weight ranged from 300 g to 330 g, were divided into control group and simulated microgravity group randomly. After 4 weeks, using isolated arterial rings from rats, arterial dilatory responsiveness of abdominal aorta were examined in vitro. And the expression of nitric oxide synthase (NOS), including endothelial NOS (eNOS) and inducible NOS (iNOS), were observed by Western blot. RESULTS: Dilatory responses of arterial rings to L-Arginine (10(-8)-10(-3) mol/L), and Acetylcholine mol/L) were decreased in simulated microgravity rats compared with that of controls; but dilatory responses of isolated aortic rings to sodium nitroprusside (mol/L) and 8-bromo-cGMP(mol/L) were similar in both simulated microgravity rats and control rats. The expression of both eNOS and iNOS had not showed significant differences between two groups. CONCLUSION: The data indicate that endothelium dependent vasorelaxation in abdominal aortic rings are decreased by 4-week simulated microgravity, and this change may be result from altered NOS activity in endothelium.


Subject(s)
Aorta, Abdominal/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/metabolism , Weightlessness Simulation , Animals , Arginine/metabolism , Cyclic GMP/metabolism , Male , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley
18.
Article in English | MEDLINE | ID: mdl-17282344

ABSTRACT

The aim of the present study was to clarify whether simulated microgravity-induced post-suspension cardiovascular deconditioning in rats could be prevented by daily short-duration standing (STD). Three groups of rats were used as subjects to perform the experiments. Compared to a control group of male Sprague-Dawley rats (CON), a group of rats with tail-suspension (SUS) for 28 d was used to simulate cardiovascular deconditioning due to microgravity. Another group of tailed-suspended rats with daily STD for 1 h was used to provide -GX (dorso-ventral) gravitational loading as countermeasure. In addition to hemodynamic changes to head-up tilt, blood pressure variability (BPV) signals were also analyzed by spectral and nonlinear analysis. The results showed 1) Blood pressure immediately decreased after head-up tilt in all three groups. After several minutes, blood pressure could restore to the initial condition in both CON and SUS+STD group, but the recovery process was slower in the latter group. In contrast, the recovery process was the slowest in the SUS group and could not restore to the initial level completely. 2) In frequency domain, power spectra has similar pattern in CON and SUS+STD group. However, they are quite different in the SUS group in that spectra peak is obvious increased in very low frequency and spectra is narrower and higher in high frequency.

19.
Zhonghua Jie He He Hu Xi Za Zhi ; 27(2): 108-11, 2004 Feb.
Article in Chinese | MEDLINE | ID: mdl-14990186

ABSTRACT

OBJECTIVE: To evaluate the inhibiting effect of niflumic acid (NFA), an inhibitor of calcium-activated chloride channel (ClCa) on airway epithelium, on the airway hyperresponsiveness in asthmatic mice. METHODS: BALB/c mice were randomly divided into an asthma group (A group), a NFA prevention asthmatic group (B group) and a sham-challenged group (C group). The airway pressure time index (APTI) and the content of ET-1 and NO in bronchoalveolar lavage fluid (BALF) in all groups were measured. With the isolated tracheal rings with integral epithelium or epithelium removed from the asthma group (A(1) group and A(2) group) and the sham-challenged group (C(1) group and C(2) group), the contractile responsiveness of various rings to methacholine (mACh) was examined, and its change was observed when the rings were exposed to NFA beforehand. RESULTS: Compared with A group (1.62 +/- 0.14), the APTI in B group (1.21 +/- 0.07) was reduced remarkably (P < 0.01), and the contents of ET-1 [(103 +/- 9) ng/L] and NO [(48.5 +/- 3.2) micromol/L] in BALF of A group were significantly higher than those in B group, [(53 +/- 5) ng/L, (23.7 +/- 2.5) micromol/L (P < 0.01), respectively]. The ratios of maximum contractility in A(1), A(2), C(1) and C(2) groups were (3.79 +/- 0.44), (2.15 +/- 0.21), (1.26 +/- 0.14) and (2.06 +/- 0.18), respectively. The contractility of A(1) group was highest among all groups (all P < 0.01), but could be effectively decreased by NFA. CONCLUSIONS: By inhibiting the special ClCa on the airway epithelium, NFA can inhibit the production of ET-1 and NO by epithelium and thus exert preventive effect on airway hyperresponsiveness in asthma.


Subject(s)
Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Niflumic Acid/therapeutic use , Animals , Bronchoalveolar Lavage Fluid/chemistry , Endothelin-1/analysis , In Vitro Techniques , Male , Mice , Nitric Oxide/analysis , Trachea/drug effects
20.
J Appl Physiol (1985) ; 95(1): 207-18, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12794097

ABSTRACT

This study was designed to compare the effectiveness of daily short-duration -Gx gravity exposure in preventing adverse changes in skeletal and cardiac muscles and bone due to simulated microgravity. Tail suspension for 28 days was used to simulate microgravity-induced deconditioning effects. Daily standing (STD) at 1 G for 1, 2, or 4 h/day or centrifugation (CEN) at 1.5 or 2.6 G for 1 h/day was used to provide -Gx gravitation as a countermeasure. The results indicate that the minimum gravity exposure requirements vary greatly in different systems. Cardiac muscle is most responsive to such treatment: 1 h/day of -Gx gravitation by STD was sufficient to prevent adverse changes in myocardial contractility; bone is most resistant: 4 h/day of -Gx gravitation only partially alleviated the adverse changes in physical and mechanical properties of the femur. The responsiveness of skeletal muscle is moderate: 4 h/day of -Gx gravitation prevented mass reduction and histomorphometric changes in the soleus muscle during a 28-day simulation period. Increasing gravitational intensity to 2.6 G showed less benefit or no additional benefit in preventing adverse changes in muscle and bone. The present work suggests that system specificity in responsiveness to intermittent gravity exposure should be considered one of the prerequisites in proposing intermittent artificial gravity as a potential countermeasure.


Subject(s)
Hypergravity , Hypogravity , Physical Conditioning, Animal/physiology , Adrenal Glands/physiology , Animals , Atrophy/prevention & control , Biomechanical Phenomena , Body Weight/physiology , Bone and Bones/pathology , Bone and Bones/physiology , Centrifugation , Heart/physiology , Hindlimb/physiology , Hindlimb Suspension/physiology , Image Processing, Computer-Assisted , Male , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Myocardial Contraction/physiology , Organ Size/physiology , Papillary Muscles/physiology , Rats , Rats, Sprague-Dawley , Testis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...