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










Publication year range
1.
J Cardiovasc Pharmacol ; 61(3): 258-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23403888

ABSTRACT

BACKGROUND: Caveolins (Cav) are structural proteins that insert into the plasma membrane to form caveolae that can bind molecules important in cardiac signal transduction and function. Cytochrome P450 epoxygenases can metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which have known cardioprotective effects. Subsequent metabolism of EETs by soluble epoxide hydrolase reduces the protective effect. AIMS: (1) To assess the effect of ischemia-reperfusion injury on expression and subcellular localization of caveolins. (2) To study the effect of EETs on caveolins. METHODS: Hearts from soluble epoxide hydrolase null (KO) and littermate control (WT) mice were perfused in Langendorff mode and subjected to 20 minutes ischemia followed by 40 minutes reperfusion. Immunohistochemistry, immunoblot, and electron microscopy were performed to study localization of caveolins and changes in ultrastructure. RESULTS: In WT heart, Cav-1 and Cav-3 were present in cardiomyocyte and capillary endothelial cell at baseline. After ischemia, Cav-1 but not Cav-3, disappeared from cardiomyocyte; moreover, caveolae were absent and mitochondrial cristae were damaged. Improved postischemic functional recovery observed in KO or WT hearts treated with 11,12-EET corresponded to higher Cav-1 expression and maintained caveolae structure. In addition, KO mice preserved the Cav-1 signaling after ischemia that lost in WT mice. CONCLUSIONS: Taken together, our data suggest that ischemia-reperfusion injury causes loss of Cav-1 and caveolins, and EETs-mediated cardioprotection involves preservation of Cav-1.


Subject(s)
Caveolae/metabolism , Caveolin 1/metabolism , Caveolin 3/metabolism , Eicosanoids/metabolism , Epoxide Hydrolases/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Biological Transport , Blotting, Western , Caveolae/ultrastructure , Epoxide Hydrolases/genetics , Heart/physiopathology , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/ultrastructure , Perfusion , Signal Transduction
2.
Am J Physiol Heart Circ Physiol ; 299(4): H1183-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20693397

ABSTRACT

Matrix metalloproteinases (MMPs) are known to degrade components of the extracellular matrix. More recently, in myocardial oxidative stress injury including ischemia-reperfusion, MMP-2 is activated and degrades troponin I and α-actinin. MMP activity is regulated at several levels. We recently showed that MMP-2 is localized in the caveolae of cardiomyocytes and is negatively regulated by caveolin-1 (Cav-1). The caveolin scaffolding domain of Cav-1 inhibits MMP-2 proteolytic activity in vitro, and Cav-1(-/-) mouse hearts have increased MMP-2 activity compared with controls. Whether this increase in MMP-2 activity translates to impaired cardiac function is unknown. Hearts isolated from Cav-1(-/-) mice and their wild-type controls were perfused as isolated working hearts and physiologically challenged with increasing increments of left atrial preload (7-22.5 mmHg). The hearts were then pharmacologically challenged with increasing concentrations of isoproterenol (0.1-100 nM). Functionally, the Cav-1(-/-) hearts were similar to the controls in heart rate, peak systolic pressure, developed pressure, and rate pressure product. At higher preload pressures, the Cav-1(-/-) hearts outperformed the control hearts. Coronary flow was significantly higher in Cav-1(-/-) hearts under all conditions. The highest concentration of isoproternol increased the heart rate of Cav-1(-/-) hearts more than in controls. Western blot analysis revealed no significant changes in troponin I or α-actinin between Cav-1(-/-) hearts and their controls. There was a significant loss of MMP-2 from both knockout and control hearts during the perfusion. In summary, despite the loss of Cav-1, Cav-1(-/-) hearts show similar or better cardiac function compared with wild-type hearts following physiological challenge or ß-adrenergic stimulation in vitro, and this appears unrelated to changes in MMP-2.


Subject(s)
Cardiac Output/physiology , Caveolin 1/deficiency , Heart Rate/physiology , Heart/physiology , Actinin/metabolism , Animals , Cardiac Output/drug effects , Cardiotonic Agents/pharmacology , Caveolin 1/genetics , Caveolin 1/metabolism , Dose-Response Relationship, Drug , Heart/drug effects , Heart Rate/drug effects , Isoproterenol/pharmacology , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Knockout , Models, Animal , Myocardium/metabolism , Troponin I/metabolism
3.
Can J Physiol Pharmacol ; 88(1): 73-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20130741

ABSTRACT

Whether ventricular cardiac myocytes of mouse contain caveolin-1 is disputed. It has been claimed to be exclusively in nearby endothelial cell profiles. Recently, matrix metalloproteinase-2 (MMP-2) was reported to be present in mouse ventricular cardiac myocytes, colocalized with caveolin-1, and caveolin-1 knockout was found to cause the loss of MMP-2 from mouse ventricular cardiac myocytes and affect their functioning. To resolve this dispute, we labeled cardiac myocytes with caveolin-1 and endothelial cells with caveolin-2. Caveolin-2 is agreed to be present exclusively in endothelial cells. The results showed that mouse ventricular myocytes were labeled with caveolin-1 antibodies independently of any caveolin-2 labeling, and endothelial cells were labeled with both caveolin-1 and caveolin-2 antibodies. This confirms that caveolin-1 is present in mouse ventricular cardiac myocytes as well as endothelial cells. Previous evidence confirms that loss of caveolin-1 affects the function of mouse ventricular cardiac myocytes and suggests that MMP-2 may be involved.


Subject(s)
Caveolin 1/physiology , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/physiology , Animals , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 2/analysis , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Heart Ventricles/chemistry , Heart Ventricles/cytology , Male , Mice , Mice, Knockout , Myocytes, Cardiac/cytology
4.
Am J Physiol Heart Circ Physiol ; 298(1): H45-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19837953

ABSTRACT

Persistent arterial hypotension is a hallmark of sepsis and is believed to be caused, at least in part, by excess nitric oxide (NO). NO can combine with superoxide to produce peroxynitrite, which activates matrix metalloproteinases (MMPs). Whether MMP inhibition in vivo protects against vascular hyporeactivity induced by endotoxemia is unknown. Male Sprague-Dawley rats were administered either bacterial lipopolysaccharide (LPS, 4 mg/kg ip) or vehicle (pyrogen-free water). Later (30 min), animals received the MMP inhibitor doxycycline (4 mg/kg ip) or vehicle (pyrogen-free water). After LPS injection (6 h), animals were killed, and aortas were excised. Aortic rings were mounted in organ baths, and contractile responses to phenylephrine or KCl were measured. Aortas and plasma were examined for MMP activity by gelatin zymography. Aortic MMP and inducible nitric oxide synthase (iNOS) were examined by immunoblot and/or immunohistochemistry. Doxycycline prevented the LPS-induced development of ex vivo vascular hyporeactivity to phenylephrine and KCl. iNOS protein was significantly upregulated in aortic homogenates from endotoxemic rats; doxycycline did not alter its level. MMP-9 activity was undetectable in aortic homogenates from LPS-treated rats but significantly upregulated in the plasma; this was attenuated by doxycycline. Plasma MMP-2 activities were unchanged by LPS. Specific MMP-2 activity was increased in aortas from LPS-treated rats. This study demonstrates the in vivo protective effect of the MMP inhibitor doxycycline against the development of vascular hyporeactivity in endotoxemic rats.


Subject(s)
Doxycycline/therapeutic use , Endotoxemia/drug therapy , Endotoxemia/physiopathology , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/therapeutic use , Animals , Aorta, Thoracic/drug effects , Blood Vessels/physiopathology , Blotting, Western , Endotoxemia/enzymology , Gelatin/metabolism , In Vitro Techniques , Lipopolysaccharides/toxicity , Male , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/metabolism , Microscopy, Confocal , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Synthase Type II/metabolism , Phenylephrine/pharmacology , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology
5.
Can J Physiol Pharmacol ; 87(3): 180-95, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19295659

ABSTRACT

Some receptors and signaling molecules, such as Rho-kinase (ROCK), localize in caveolae. We asked whether the function of histamine receptors (H(1)) and 5-hydroxytryptamine (serotonin) receptors (5-HT(2A)) in bovine tracheal smooth muscle are modified after caveolae disruption and if so, whether the altered ROCK activity plays a role in this modification. Methyl-beta-cyclodextrin (MbetaCD), used to deplete membrane cholesterol, was shown to disrupt caveolae and diminish sustained contractions to histamine (approximately 80%), 5-HT (100%), alpha-methyl-5-HT (100%), and KCl (approximately 30%). Cholesterol-loaded MbetaCD (CL-MbetaCD) restored the responses to KCl and partially restored the responses to agonists. ROCK inhibition by Y-27632 diminished contractions to histamine (approximately 85%) and 5-HT (approximately 59%). 5-HT or histamine stimulation augmented ROCK activity. These increases were reduced by MbetaCD and partially reestablished by CL-MbetaCD. The increase in intracellular Ca(2+) that was induced by both agonists was reduced by MbetaCD. The presence of caveolin-1 (Cav-1), H1, 5-HT(2A), and ROCK1 was corroborated by immunoblotting of membrane fractions from sucrose gradients and by confocal microscopy. H(1) receptors coimmunoprecipitated with Cav-1 in caveolar and noncaveolar membrane fractions, whereas 5-HT(2A) receptors appeared to be restricted to noncaveolar membrane fractions. We conclude that caveolar and cholesterol integrity are indispensable for the proper functionality of the H(1) and 5-HT(2A) receptors through their Rho/ROCK signaling.


Subject(s)
Caveolae/physiology , Cholesterol/physiology , Membrane Lipids/physiology , Muscle, Smooth/physiology , Receptor, Serotonin, 5-HT2A/physiology , Receptors, Histamine H1/physiology , Trachea/physiology , rho-Associated Kinases/physiology , Animals , Calcium/metabolism , Cattle , Caveolin 1/physiology , Signal Transduction , beta-Cyclodextrins/pharmacology
6.
J Cell Mol Med ; 13(2): 352-64, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19166483

ABSTRACT

In mouse intestine, caveolae and caveolin-1 (Cav-1) are present in smooth muscle (responsible for executing contractions) and in interstitial cells of Cajal (ICC; responsible for pacing contractions). We found that a number of calcium handling/dependent molecules are associated with caveolae, including L-type Ca(2+) channels, Na(+)-Ca(2+) exchanger type 1 (NCX1), plasma membrane Ca(2+) pumps and neural nitric oxide synthase (nNOS), and that caveolae are close to the peripheral endo-sarcoplasmic reticulum (ER-SR). Also we found that this assemblage may account for recycling of calcium from caveolar domains to SR through L-type Ca (+) channels to sustain pacing and contractions. Here we test this hypothesis further comparing pacing and contractions under various conditions in longitudinal muscle of Cav-1 knockout mice (lacking caveolae) and in their genetic controls. We used a procedure in which pacing frequencies (indicative of functioning of ICC) and contraction amplitudes (indicative of functioning of smooth muscle) were studied in calcium-free media with 100 mM ethylene glycol tetra-acetic acid (EGTA). The absence of caveolae in ICC inhibited the ability of ICC to maintain frequencies of contraction in the calcium-free medium by reducing recycling of calcium from caveolar plasma membrane to SR when the calcium stores were initially full. This recycling to ICC involved primarily L-type Ca(2+) channels; i.e. pacing frequencies were enhanced by opening and inhibited by closing these channels. However, when these stores were depleted by block of the sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) pump or calcium release was activated by carbachol, the absence of Cav-1 or caveolae had little or no effect. The absence of caveolae had little impact on contraction amplitudes, indicative of recycling of calcium to SR in smooth muscle. However, the absence of caveolae slowed the rate of loss of calcium from SR under some conditions in both ICC and smooth muscle, which may reflect the loss of proximity to store operated Ca channels. We found evidence that these channels were associated with Cav-1. These changes were all consistent with the hypothesis that a reduction of the extracellular calcium associated with caveolae in ICC of the myenteric plexus, the state of L-type Ca(2+) channels or an increase in the distance between caveolae and SR affected calcium handling.


Subject(s)
Calcium/metabolism , Caveolae/metabolism , Caveolin 1/metabolism , Intestinal Mucosa , Intestines , Muscle Contraction/physiology , Muscle, Smooth/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , Animals , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/metabolism , Calcium-Transporting ATPases/metabolism , Caveolin 1/genetics , Chelating Agents/metabolism , Egtazic Acid/metabolism , Intestinal Mucosa/metabolism , Intestines/anatomy & histology , Male , Mice , Mice, Knockout , Nicardipine/metabolism , Sarcoplasmic Reticulum/metabolism
7.
Eur J Pharmacol ; 591(1-3): 80-7, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18634779

ABSTRACT

Plasma membrane calcium ATPase (PMCA) is an important calcium extrusion mechanism in smooth muscle cells. PMCA4 is the predominant isoform operating in conditions of high intracellular calcium during contraction. PMCA appears to be localized in lipid rafts and caveolae. In this study we examined the effects of the PMCA4-selective inhibitor caloxin 1c2 (5 microM) in intestine of caveolin-1 knockout mice and in bovine tracheal smooth muscle after caveolae disruption on PMCA4 function. Small intestinal tissues from control mice treated with caloxin 1c2 showed a higher contractile response of the longitudinal smooth muscle to Carbachol (10 microM) when compared to control tissues treated with a similar concentration of a control peptide. This effect of caloxin 1c2 was not found in tissues from caveolin-1 knockout mice. Immunohistochemistry and Western blotting of membrane fractions showed that PMCA was co-localized with caveolin-1 in smooth muscle plasma membrane in control tissues. One of the PMCA4 splice variant bands was missing in the lipid raft-enriched fraction prepared from caveolin-1 knockout tissue. In bovine tracheal smooth muscle tissue, caveolae disruption by cholesterol depletion led to the diminution of caveolin-1 and PMCA4b immunoreactivities, previously co-localized in the smooth muscle plasma membrane, and to the loss of the increase in Carbachol-induced contraction by caloxin 1c2. Our results suggest that the calcium removal function of PMCA4 in smooth muscle cells is dependent on its presence in intact caveolae. We suggest that this is due to the close spatial arrangement that allows calcium extrusion from a privileged cytosolic space between caveolae and sarcoplasmic reticulum.


Subject(s)
Calcium/metabolism , Caveolin 1/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Carbachol/pharmacology , Cattle , Caveolae/metabolism , Caveolin 1/genetics , Cytosol/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Male , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Peptides/pharmacology , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Trachea/drug effects , Trachea/metabolism
8.
J Cell Mol Med ; 12(4): 1404-15, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18400048

ABSTRACT

Neuronal nitric oxide synthase (nNOS) in myenteric neurons is activated during peristalsis to produce nitric oxide which relaxes intestinal smooth muscle. A putative nNOS is also found in the membrane of intestinal smooth muscle cells in mouse and dog. In this study we studied the possible functions of this nNOS expressed in mouse small intestinal smooth muscle colocalized with caveolin-1(Cav-1). Cav-1 knockout mice lacked nNOS in smooth muscle and provided control tissues. 60 mM KCl was used to increase intracellular [Ca(2+)] through L-type Ca(2+) channel opening and stimulate smooth muscle NOS activity in intestinal tissue segments. An additional contractile response to LNNA (100 microM, NOS inhibitor) was observed in KCl-contracted tissues from control mice and was almost absent in tissues from Cav-1 knockout mice. Disruption of caveolae with 40 mM methyl-beta cyclodextrin in tissues from control mice led to the loss of Cav-1 and nNOS immunoreactivity from smooth muscle as shown by immunohistochemistry and a reduction in the response of these tissues to N-omega-nitro-L-arginine (LNNA). Reconstitution of membrane cholesterol using water soluble cholesterol in the depleted segments restored the immunoreactivity and the response to LNNA added after KCl. Nicardipine (1 microM) blocked the responses to KCl and LNNA confirming the role of L-type Ca(2+) channels. ODQ (1 microM, soluble guanylate cyclase inhibitor) had the same effect as inhibition of NOS following KCl. We conclude that the activation of nNOS, localized in smooth muscle caveolae, by calcium entering through L-type calcium channels triggers nitric oxide production which modulates muscle contraction by a cGMP-dependent mechanism.


Subject(s)
Caveolin 1/metabolism , Intestine, Small/enzymology , Muscle Contraction/physiology , Muscle, Smooth/enzymology , Nitric Oxide Synthase Type I/metabolism , Animals , Blotting, Western , Calcium/metabolism , Cholesterol/deficiency , Cryoultramicrotomy , Immunohistochemistry , Immunoprecipitation , Intestine, Small/cytology , Intestine, Small/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Nitric Oxide/metabolism , Nitroarginine/pharmacology , Protein Transport/drug effects
9.
J Cell Mol Med ; 11(5): 1069-86, 2007.
Article in English | MEDLINE | ID: mdl-17979883

ABSTRACT

Matrix metalloproteinase-2 (MMP-2) may play roles at intracellular and extracellular sites of the heart in ischaemia/reperfusion injury. Caveolins (Cav-1, -2 and -3) are lipid raft proteins which play roles in cell sig-nalling. This study examined, using immunohistochemistry and two photon confocal microscopy, if MMP-2 and caveolins co-localize at the plasma membrane of cardiac cells: cardiomyocytes (CM), fibroblasts (FB) and capillary endothelial cells (CEC) in the left ventricle (LV) of the Cav-1(+/+) and Cav-1(-/-) mouse heart. In Cav-1(+/+) mouse LV MMP-2 and Cav-1 co-localized at CM plasma membranes, and at multiple locations in FB and CEC. MMP-2 co-localized with Cav-2 only at CEC. MMP-2 co-localized with Cav-3 at CM plasma membranes and Z-lines, and partially at FB and CEC. In Cav-1(-/-) LV Cav-1 and MMP-2 were absent or reduced everywhere. Cav-2 appeared at CEC despite the absence of Cav-1. Cav-3 appeared at CM plasma membranes and Z-lines, FB and CEC. Also, FAK in FB and c-Kit in interstitial Cajal-like cells (ICLC) were completely absent. By transmission electron microscopy in Cav-1(+/+), regular size caveolae (Cav) were at CEC, irregular size Cav were at CM and a few were at FB. In Cav-1(-/-) there were few Cav at CM and FB and some at CEC. To conclude, MMP-2 is closely associated with caveolins at FB and CEC as well as at CM. Also, MMP-2 is closely associated with FAK at FB and c-Kit at ICLC. Thus, Cav-1 expression is not necessary for Cav-2 expression. Cav-3 or Cav-3 with Cav-2 has the capability to make Cav.


Subject(s)
Caveolins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , Myocardium/cytology , Myocardium/enzymology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Caveolin 1/metabolism , Caveolin 2/metabolism , Caveolin 3/metabolism , Discoidin Domain Receptors , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/ultrastructure , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/ultrastructure , Male , Mice , Myocardium/ultrastructure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Protein Isoforms/metabolism , Protein Transport , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , von Willebrand Factor/metabolism
10.
Auton Neurosci ; 131(1-2): 36-44, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-16844426

ABSTRACT

We examined the inhibitory mediators acting on each of the longitudinal (LM) and circular muscle (CM) layers of mouse small intestine in the presence of atropine, prazosin and timolol. Nitric oxide (NO) and apamin-sensitive mediators exerted an inhibitory tone on pacing frequency in CM, observed as an increased frequency upon treatment with N-omega-nitro-l-arginine (LNNA) or apamin. This effect was not seen in LM. 1H-(1,2,4)oxadiazolo(4,3-A)quinazoline-1-one (ODQ) abolished the relaxation in response to electric field stimulation (EFS) in LM in a manner similar to LNNA indicating that the inhibitory mediator in this layer in NO acting via soluble guanylate cyclase. On the other hand, in CM neither LNNA nor apamin was capable of reducing the inhibition in response to EFS and their combination left a residual relaxation of 25%. ODQ reduced the EFS-evoked relaxation more effectively than LNNA at higher frequencies indicating that another ODQ-sensitive mediator was active in CM. ODQ also blocked the relaxation to exogenous vasoactive intestinal peptide in CM. In LM, the relaxation due to sodium nitroprusside was equally blocked by ODQ and apamin, while in CM, its effects were only reduced by ODQ and not apamin. These results indicate that there are differences in the inhibitory mediators and the mechanisms of action involved in LM and CM relaxation.


Subject(s)
Intestine, Small/innervation , Muscle, Smooth/physiology , Animals , Apamin/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Evoked Potentials/drug effects , Evoked Potentials/physiology , Evoked Potentials/radiation effects , In Vitro Techniques , Intestine, Small/physiology , Male , Mice , Mice, Inbred BALB C , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Contraction/radiation effects , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle Relaxation/radiation effects , Muscle, Smooth/drug effects , Muscle, Smooth/innervation , Nitric Oxide Donors/pharmacology , Nitroarginine/pharmacology , Nitroprusside/pharmacology , Oxadiazoles/pharmacology
11.
Am J Physiol Gastrointest Liver Physiol ; 291(6): G1020-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16782699

ABSTRACT

beta-Adrenoceptors are G protein-coupled receptors whose functions are closely associated with caveolae in the heart and cultured cell lines. In the gut, they are responsible, at least in part, for the mediation of the sympathetic stimulation that might lead to intestinal paralysis postoperatively. We examined the effect of caveolin-1 knockout on the beta-adrenoceptor response in mouse small intestine. The relaxation response to (-)-isoprenaline in carbachol-contracted small intestinal tissue segments was reduced in caveolin-1 knockout mice (cav1(-/-)) compared with their genetic controls (cav1(+/+)). Immunohistochemical staining showed that beta-adrenoceptor expression was similar in both strains in gut smooth muscle. Selective beta-adrenoceptor blockers shifted the concentration response curve (CRC) of (-)-isoprenaline to the right in cav1(+/+) intestine, but not in cav1(-/-), with greatest shift in case of the beta(3)-blocker, SR59230A. The CRC of the selective beta(3)-agonist BRL 37344 was also shifted to the right in cav1(-/-) compared with cav1(+/+). The cAMP-dependent protein kinase (PKA) inhibitor H-89 shifted the CRC of (-)-isoprenaline to the right in cav1(+/+) but not in cav1(-/-). H-89 reduced the relaxation due to forskolin and dibutyryl cAMP in cav1(+/+) but not in cav1(-/-), suggesting a reduction in PKA activity in cav1(-/-). In cav1(+/+), PKA was colocalized with caveolin-1 in the cell membrane, but PKA immunoreactivity persisted in cav1(-/-). Examination of PKA expression in the lipid raft-rich membrane fraction of the jejunum revealed reduced PKA expression in cav1(-/-) compared with cav1(+/+). The results of the present study show that the function of beta-adrenoceptors is reduced in cav1(-/-) small intestine likely owing to reduced PKA activity.


Subject(s)
Caveolin 1/metabolism , Intestine, Small/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction/physiology , Animals , Caveolin 1/genetics , Male , Mice , Mice, Knockout
13.
Am J Physiol Gastrointest Liver Physiol ; 290(2): G394-403, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16166342

ABSTRACT

Recently, we showed that caveolin-1 (cav1) knockout mice (Cav1(-/-) mice) have impaired nitric oxide (NO) function in the longitudinal muscle (LM) layer of the small intestine. The defect was a reduced responsiveness of the muscles to NO compensated by an increase in the function of apamin-sensitive, nonadrenergic, noncholinergic (NANC) mediators. In the present study, we examined similarly the effects of cav1 knockout on the relaxation in circular muscle (CM) of the mouse small intestine. CM of Cav1(-/-) mice also showed defective NO function, but less than in LM, as well as more activation of apamin-sensitive NANC mediators. CM of Cav1(-/-) mice, like LM, lacked cav1 but retained small amounts of cav3 and caveolae in the outer CM layer. In addition, we also examined the effects of a soluble guanylate cyclase inhibitor, 1H-[1,2,4]oxadiazolo-[4,3-alpha]quinazolin-1-one (ODQ), on electric field stimulation (EFS)-mediated relaxation in both LM and CM. ODQ had an effect similar to the block of NO synthesis. Moreover, we compared the actions of two NO donors in the LM and CM of control and Cav1(-/-) mice. Similar to LM, CM of Cav1(-/-) mice showed a reduced responsiveness to the NO donors sodium nitroprusside and S-nitroso-N-acetyl penicillamine. However, both ODQ and apamin blocked the inhibitory effects of the NO donors in LM, whereas apamin had no effect in CM. In conclusion, cav1 knockout affects NO function in both LM and CM, but its effects in CM differ significantly.


Subject(s)
Autonomic Nervous System/physiology , Caveolin 1/genetics , Caveolin 1/physiology , Muscle, Smooth/physiology , Animals , Apamin/pharmacology , Immunohistochemistry , In Vitro Techniques , Jejunum/drug effects , Jejunum/innervation , Jejunum/ultrastructure , Male , Mice , Mice, Knockout , Microscopy, Electron , Muscle Relaxation/physiology , Muscle, Smooth/innervation , Muscle, Smooth/ultrastructure , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Nitroarginine/pharmacology , Nitroprusside/pharmacology , Oxadiazoles/pharmacology , Quinoxalines/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Synaptic Transmission/physiology , Tetrodotoxin/pharmacology
14.
Histochem Cell Biol ; 126(1): 9-16, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16369777

ABSTRACT

Confocal microscopic images were obtained from the immunohistochemical sections of jejeunum to determine the localization/colocalization between caveolin-1, caveolin-2 and caveolin-3 in intestinal smooth muscle cells (SMCs) and interstitial cells of Cajal (ICC) of Cav1(+/+) and Cav1(-/-) mouse. Intestinal regions were segmented [inner circular muscle (icm), outer circular muscle (ocm), myenteric plexus region (mp), and longitudinal muscle (lm)] by LSM 5 and analyzed by ImageJ to show Pearson's correlation (r (p)) and overlap coefficient (r) of colocalization. In the intestine of Cav1(+/+), caveolin-1 (cav1) was colocalized with caveolin-2 (cav2) and caveolin-3 (cav3). Cav2 also was well colocalized with cav3. In the intestine of Cav1(-/-), cav1 and cav2 were absent in all images, but reduced cav3 was expressed in ocm. Caveolae were present in cell types with cav1 in Cav1(+/+), and present with cav3 in ocm of Cav1(-/-). C-kit occurred in deep muscular plexus (ICC-DMP) and myenteric plexus (ICC-MP), in both Cav1(+/+) and Cav1(-/-), and colocalized with cav1 and cav2 in the intestine of Cav1(+/+). Cav3 was absent/present at low immunoreactivity in ICC-DMP and ICC-MP of the intestines of Cav1(+/+) and Cav1(-/-). To conclude, cav1 is necessary for the expression of cav2 in SMC and ICC of intestine and facilitates, but is not necessary for the expression of cav3.


Subject(s)
Caveolins/genetics , Caveolins/metabolism , Intestinal Mucosa/metabolism , Muscle, Smooth/metabolism , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Caveolin 2/genetics , Caveolin 2/metabolism , Caveolin 3/genetics , Caveolin 3/metabolism , Fluorescent Antibody Technique, Indirect , Image Processing, Computer-Assisted , Intestines/cytology , Intestines/ultrastructure , Isomerism , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Muscle, Smooth/cytology , Muscle, Smooth/ultrastructure
15.
Can J Physiol Pharmacol ; 83(8-9): 657-63, 2005.
Article in English | MEDLINE | ID: mdl-16333367

ABSTRACT

After 58 years in science, mostly in pharmacology, one gains perspective. Mine is that there have been important changes over this time, some good and some questionable. In this commentary, I try to reveal how I got to this stage, partially explaining my biases, and possibly helping others learn from my experiences including mistakes. Changing from seeking an M.D. to cellular biology and then to pharmacology early in my career were the best moves I made. The next best move was migration to Canada, away from the McCarthy-McCarran hysteria. Arriving at a time after the end of World War II when science in Canada was expanding was very good luck. I had an excellent opportunity to enjoy both the administration (as Chair of the first independent Department of Pharmacology at the University of Alberta) and the practice of pharmacology (as a practitioner of research on smooth muscle in health and disease). For me, the practice of research has always won over administration when a choice had to be made. Early on, I began to ask questions about educational practices and tried to evaluate them. This led me to initiate changes in laboratories and to seek nondidactic educational approaches such as problem-based learning. I also developed questions about the practice of anonymous peer review. After moving to McMaster in 1975, I was compelled to find a solution for a failed "Pharmacology Program" and eventually developed the first "Smooth Muscle Research Program". Although that was a good solution for the research component, it did not solve the educational needs. This led to the development of "therapeutic problems", which were used to help McMaster medical students educate themselves about applied pharmacology. Now these problems are being used to educate pharmacology honours and graduate students at the University of Alberta. The best part of all these activities is the colleagues and friends that I have interacted with and learned from over the years, and the realization that many of them have collaborated with me again in this volume.


Subject(s)
Pharmacology/education , Canada , History, 20th Century , Muscle, Smooth , Pharmacology/history , Research , Teaching , Universities
16.
Br J Pharmacol ; 145(8): 1017-26, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15937515

ABSTRACT

Caveolin-1 is a plasma membrane-associated protein that is responsible for caveolae formation. It plays an important role in the regulation of the function of different signaling molecules, among which are the different isoforms of nitric oxide synthase (NOS). Nitric oxide (NO) is known to be an important inhibitory mediator in the mouse gut. Caveolin-1 knockout mice (Cav1(-/-)) were used to examine the effect of caveolin-1 absence on the NO function in the mouse small intestine (ileum and jejunum) compared to their genetic controls and BALB/c controls. Immunohistochemical staining showed loss of caveolin-1 and NOS in the jejunal smooth muscles and myenteric plexus interstitial cells of Cajal (ICC) of Cav1(-/-) mice; however, nNOS immunoreactive nerves were still present in myenteric ganglia. Under nonadrenergic noncholinergic (NANC) conditions, small intestinal tissues from Cav1(-/-) mice relaxed to electrical field stimulation (EFS), as did tissues from control mice. Relaxation of tissues from control mice was markedly reduced by N-omega-nitro-L-arginine (10(-4) M), but relaxation of Cav1(-/-) animals was affected much less. Also, Cav1(-/-) mice tissues showed reduced relaxation responses to sodium nitroprusside (100 microM) compared to controls; yet there were no significant differences in the relaxation responses to 8-bromoguanosine-3': 5'-cyclic monophosphate (100 microM). Apamin (10(-6) M) significantly reduced relaxations to EFS in NANC conditions in Cav1(-/-) mice, but not in controls. The data from this study suggest that caveolin-1 gene knockout causes alterations in the smooth muscles and the ICC, leading to an impaired NO function in the mouse small intestine that could possibly be compensated by apamin-sensitive inhibitory mediators.


Subject(s)
Ileum/physiology , Jejunum/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Nitric Oxide/physiology , Animals , Electric Stimulation , Enzyme Inhibitors/pharmacology , Ileum/metabolism , Ileum/ultrastructure , Immunohistochemistry , In Vitro Techniques , Jejunum/metabolism , Jejunum/ultrastructure , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Muscle, Smooth/ultrastructure , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology
17.
Am J Physiol Gastrointest Liver Physiol ; 286(2): G234-43, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14715518

ABSTRACT

Activation of a two-dimensional sheet network (5 parallel chains of 5 cells each) of simulated intestinal smooth muscle cells (SMCs) by one interstitial cell of Cajal (ICC) was modeled by PSpice simulation. The network of 25 cells was not interconnected by gap-junction channels; instead, excitation was transmitted by the electric field that develops in the junctional clefts (JC) when the prejunctional membrane fires an action potential (AP). Transverse propagation between the parallel chains occurs similarly. The ICC cell was connected to cell E5 of the network [5th cell of the 5th (E) chain] via a high-resistance junction. The stimulating current, applied to the ICC cell interior, was made to resemble the endogenous undershooting slow wave (I(SW)). An I(SW) of 2.4 nA (over a rise time of 4 ms) took the ICC cell from a resting potential (RP) of -80 mV to a membrane potential of -41 mV. The slow wave produced a large negative cleft potential in the JC (V(JC); ICC-E5). The V(jc) brought the postjunctional membrane of E5 to threshold, causing this cell to fire an AP. This, in turn, propagated throughout the SMC network. If the ICC cell was given an RP of -55 mV (like SMC) and a slow wave of 40 mV amplitude (I(SW) of 1.8 nA), it still activated the SMC network. This was also true when the ICC cell was made excitable (developing an overshooting, fast-rising AP). In summary, one ICC cell displaying a slow wave was capable of activating a network of SMC in the absence of gap junctions.


Subject(s)
Computer Simulation , Intestines/physiology , Models, Biological , Myocytes, Smooth Muscle/physiology , Animals , Electric Stimulation , Electrophysiology , Humans , Intestines/cytology
18.
Biochim Biophys Acta ; 1589(2): 93-103, 2002 Apr 03.
Article in English | MEDLINE | ID: mdl-12007785

ABSTRACT

Changes in endothelial cell (EC) shape result in inter-EC gap formation and subsequently regulate transendothelial passage. In this work, we investigated the effects of protein phosphorylation (induced by inhibition of protein phosphatases) on EC shape changes. Treatment of bovine pulmonary artery endothelial cells (BPAEC) with calyculin A (100 nM, an inhibitor of protein Ser/Thr phosphatases 1 and 2A) resulted in cell retraction, surface bleb formation and cell rounding. Trypan blue and electrophysiological experiments suggested that the plasma membrane of these rounded cells maintained functional integrity. Calyculin A-induced morphological changes were strongly inhibited by staurosporine, but not affected by specific inhibitors of the myosin light chain (MLC) kinase, protein kinases A, C and G, and tyrosine kinases. The calyculin A effects were not mimicked by phorbol myristate acetate, dibutyryl cAMP, 8-bromo-cGMP or ionomycin. Cytochalasin B (an inhibitor of actin polymerization) almost completely abolished such shape changes while colchicine (an inhibitor of microtubule polymerization) had no inhibitory effect at all. Ca(2+) imaging experiments showed that the morphological changes were not associated with any global or local cytosolic Ca(2+) concentration ([Ca(2+)](i)) elevation. The results suggest that calyculin A unmasked the basal activities of some protein Ser/Thr kinases other than MLC kinase and protein kinases A, C and G; these unknown kinases might cause BPAEC shape changes by a mechanism involving actin polymerization but not [Ca(2+)](i) elevation.


Subject(s)
Actins/metabolism , Calcium/metabolism , Endothelium, Vascular/drug effects , Oxazoles/pharmacology , Actins/chemistry , Calcium/analysis , Cations, Divalent , Cell Line/drug effects , Cell Size/drug effects , Colchicine/pharmacology , Cytochalasin B/pharmacology , Electrophysiology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Marine Toxins , Polymers/chemistry , Protein Kinase C/antagonists & inhibitors , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...