Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Cardiobiol ; 5(1)2017 May.
Article in English | MEDLINE | ID: mdl-30931350

ABSTRACT

AIM: The purpose of the study was to localize matrix metalloproteinase (MMP)-14, -9, and -2 in the A7r5 smooth muscle cell and to understand the interaction between these MMPs and the cytoskeleton. This interaction was observed under non-stimulating and phorbol 12, 13-dibutyrate (PDBu)-stimulating conditions. METHODS: Confocal microscopy was utilized to define the localizations of MMPs and tissue inhibitor of matrix metalloproteinases (TIMPs) in the A7r5 cell and to determine interaction between MMPs and the cytoskeleton. Under PDBu-stimulating conditions, the presence of MMP active forms and activity by gel zymography was evaluated in the A7r5 cell. Actin and microtubule-polymerization inhibitors were used to evaluate MMP interaction with the cytoskeleton and the cytoskeleton was observed on matrix and within a Type I collagen gel. RESULTS: MMP-14, -9, and -2 were localized to the podosome in the A7r5 smooth muscle cell and interactions were seen with these MMPs and the actin cytoskeleton. PDBu-stimulation induced increases in the protein abundance of the active forms of the MMPs and MMP-2 activity was increased. MMPs also interact with a-actin and not ß-tubulin in the A7r5 cell. Galardin, also known as GM-6001, was shown to inhibit podosome formation and prevented MMP localization to the podosome. This broad spectrum MMP inhibitor also prevented collagen gel contraction and prevented cell adhesion and spreading of A7r5 cells within this collagen matrix. CONCLUSION: MMPs are important in the formation and function of podosomes in the A7r5 smooth muscle cell. MMPs interact with a-actin and not ß-tubulin in the A7r5 cell. Podosomes play an important role in cell migration and understanding the function of podosomes can lead to insights into cancer metastasis and cardiovascular disease.

2.
Can J Physiol Pharmacol ; 86(7): 403-15, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18641689

ABSTRACT

Estrogen plays an important role in skeletal physiology by maintaining a remodeling balance between the activity of osteoblasts and osteoclasts. In an attempt to decipher the mechanism through which estrogen elicits its action on osteoblasts, experimentation necessitated the development of a culturing environment reduced in estrogenic compounds. The selected medium (OPTI-MEM) is enriched to sustain cultures under reduced fetal bovine serum (FBS) conditions and is devoid of the pH indicator phenol red, a suspected estrogenic agent. This protocol reduced the concentration of FBS supplementation to 0% through successive 24 h incubations with diminishing amounts of total FBS (1%, 0.1%, and 0%). The protocol does not appear to alter the viability, cell morphology, or osteoblast-like phenotype of 7F2 and UMR-106 cell lines when compared with control cells grown in various concentrations of FBS. Although the rate of mitotic divisions declined, the 7F2 and UMR-106 cultures continued to express osteoblast-specific markers and exhibited estrogen responsiveness. These experimental findings demonstrate that the culture protocol developed did not alter the osteoblast nature of the cell lines and provides a model system to study estrogen's antiresorptive role on skeletal turnover.


Subject(s)
Fetal Blood/physiology , Osteoblasts/physiology , Serum/physiology , Adaptation, Physiological , Alkaline Phosphatase/metabolism , Animals , Cattle , Cell Differentiation/physiology , Cell Line , Cell Survival/drug effects , Estradiol/pharmacology , Female , Genetic Markers , Immunohistochemistry , Mice , Osteocalcin/biosynthesis , Pregnancy , RANK Ligand/genetics , RANK Ligand/physiology , Reverse Transcriptase Polymerase Chain Reaction
3.
Can J Physiol Pharmacol ; 84(8-9): 867-75, 2006.
Article in English | MEDLINE | ID: mdl-17111031

ABSTRACT

In the present study, we investigated the reorganization of alpha- and beta-actin in the contracting A7r5 smooth muscle cell. The remodeling of these actin variants was markedly different in response to increasing concentrations of phorbol 12, 13-dibutyrate (PDBu). At the lowest concentrations (< or =10(-7) mol/L), cells showed an approximately 70% loss in alpha-actin stress fibers with robust transport of this isoform to podosomes. By comparison, beta-actin remained in stress fibers in cells stimulated at low concentrations (< or =10(-7) mol/L) of PDBu. However, at high concentrations (> or =10(-6)mol/L) approximately 50% of cells showed transport of beta-actin to podosomes. Consistent with these findings, staining with phalloidin indicated a significant decrease in the whole-cell content of F-actin with PDBu treatment. However, staining with DNase I indicated no change in the cellular content of G-actin, suggesting reduced access of phalloidin to tightly packed actin in the podosome core. Inhibition of protein kinase C (staurosporine, bisindolymaleimide) blocked PDBu-induced (5 x 10(-8) mol/L) loss in alpha-actin stress fibers or reversed podosome formation with re-establishment of alpha-actin stress fibers. By comparison, these inhibitors caused partial loss of beta-actin stress fibers. The results support our earlier conclusion of independent remodeling of alpha- and beta-actin cytoskeletal structure and suggest that the regulation of these structures is different.


Subject(s)
Actins/physiology , Myocytes, Smooth Muscle/physiology , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Maleimides/pharmacology , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Protein Isoforms/physiology , Protein Kinase C/antagonists & inhibitors , Rats , Staurosporine/pharmacology
4.
Clin Nutr ; 22(4): 359-63, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12880602

ABSTRACT

We compared the effects of fasting (36 h) on blood pressure and aortic contractile responsiveness in lean and obese Zucker rats. Fasting of lean animals resulted in a significant loss in body weight (-9.1 +/- 0.1%) and reduction in systolic blood pressure (-11.4 +/- 1.9 mmHg). Fasting plasma triacylglycerols (-76%) and beta-hydroxybutryic acid (beta-HBA) (+ 218%) were significantly decreased and increased, respectively. The fasting plasma concentrations of insulin (-72%) were significantly decreased, whereas glucose and epinephrine (Epi) were not changed in lean rats. The fasting of obese rats also resulted in weight loss (-5.6 +/- 1.3%) but did not cause a significant reduction of blood pressure. The plasma total cholesterol (+18%) was increased, triacylglycerols (-42%) were decreased and beta-HBA levels were unchanged in fasted obese rats. Similar to lean animals, the insulin levels of fasted obese rats were significantly decreased (-85%), whereas glucose and Epi concentrations were not significantly changed. Fasting of lean animals had no effect on the maximal contractile response of aortae to high K(+) and phorbol 12, 13 dibutyrate (PDBu) but significantly reduced the response to norepinephrine (NE) (% reference: fed, 61.1 +/- 11.0; fasted, 45.6 +/- 4.5). In addition, the concentration for half-maximal response (ED(50)) to NE was increased in fasted lean rats (fed, 1.8+/-0.2 x 10(-8)M; fasted, 3.0+/-0.3 x 10(-8)M). By comparison, fasting of obese rats had no significant effect on the contractile response to K(+), NE, or PDBu. The results show that short-term food withdrawal induces significant changes in vascular contractile properties of lean but not obese rats. Because fasting-induced changes were variable depending on the agonist, the results further suggest that the mechanism did not involve a general loss or enhancement in functional status.


Subject(s)
Blood Pressure/physiology , Fasting/physiology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Obesity/physiopathology , Thinness/physiopathology , 3-Hydroxybutyric Acid/blood , Animals , Aorta, Thoracic/physiology , Disease Models, Animal , Epinephrine/blood , Female , Insulin/blood , Norepinephrine/blood , Obesity/blood , Obesity/genetics , Rats , Rats, Zucker , Thinness/blood , Triglycerides/blood , Weight Loss/physiology
5.
Am J Physiol Cell Physiol ; 285(1): C76-87, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12637266

ABSTRACT

Using laser scanning confocal, fluorescence resonance energy transfer (FRET), and atomic force (AFM) microscopy, we investigated association of protein kinase C (PKC)-alpha with microtubules during stimulus-induced relocalization in A7r5 smooth muscle cells. Confocal microscopy with standard immunostaining techniques confirmed earlier observations that colchicine disruption of microtubules blocked PKC-alpha localization in the perinuclear region of the cell caused by phorbol 12,13-dibutyrate (PDBu; 10-6M). Dual immunostaining suggested colocalization of PKC-alpha and beta-tubulin in both unstimulated and PDBu-treated cells. This finding was verified by FRET microscopy, which indicated that association of PKC-alpha was heterogeneous in distribution and confined primarily to microtubules in the perinuclear region. FRET analysis further showed that association between the molecules was not lost during colchicine-induced dissolution of microtubules, suggesting formation of tubulin-PKC-alpha complexes in the cytosol. Confocal imaging indicated that perinuclear microtubular structure was more highly sensitive to colchicine dissolution than other regions of the cell. Topographic imaging of fixed cells by AFM indicated a well-defined elevated structure surrounding the nucleus that was absent in colchicine-treated cells. It was calculated that the volume of the nuclear sleevelike structure of microtubules increased approximately fivefold in PDBu-treated cells, suggesting a probable increase in microtubular mass. In light of PKC-alpha localization, increased colchicine sensitivity, and their volume change in stimulated cells, the results suggest that perinuclear microtubules form a specialized structure that may be more dynamically robust than in other regions of the cell. PKC-alpha could contribute to this dynamic activity. Alternatively, perinuclear microtubules could act as a scaffold for regulatory molecule interaction at the cell center.


Subject(s)
Microtubules/enzymology , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/metabolism , Animals , Aorta/cytology , Carcinogens/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Colchicine/pharmacology , Microscopy, Atomic Force , Microtubules/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C-alpha , Rats
6.
Acta Physiol Scand ; 177(2): 197-205, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12558556

ABSTRACT

AIM: Using confocal microscopy and standard immunohistochemical techniques, changes in the structure of alpha-actin and beta-actin as well as the distribution of myosin II were studied in the contracting A7r5 smooth muscle cell. RESULTS: Prior to stimulation, each of the three proteins were incorporated into filamentous structures extending the length of the cell body. At 30 min after stimulation by phorbol 12, 13 dibutyrate (PDBu), the system of densely packed beta-actin fibres was retained. By comparison, alpha-actin and myosin were observed to undergo significant remodelling, characterized by loss in fibre structure and the formation of brightly fluorescing peripheral bodies. Co-immunoprecipitation of alpha-actin and myosin II suggested an association between the proteins. Consistent with this observation, dual immunostaining for alpha-actin and myosin revealed strong co-localization of the two proteins prior to stimulation. Following PDBu stimulation myosin II was observed to partially disassociate from alpha-actin structure but showed significant co-localization with alpha-actin filaments and peripheral bodies throughout the interval of contraction. The use of cytochalasin B to block actin polymerization or the selective dissolution of alpha-actin cable structure by thapsigargin produced similar patterns of change in alpha-actin structure and the localization of myosin II. CONCLUSION: The results support the concept of myosin liability and potential for remodelling. The results suggest that myosin undergoes extensive relocalization in association with alpha-actin remodelling which may be an important determinant of contractile function in the A7r5 smooth muscle cell.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth/physiology , Myosin Type II/physiology , Actins/analysis , Actins/physiology , Animals , Cells, Cultured , Cytochalasin B/pharmacology , Enzyme Inhibitors/pharmacology , Immunohistochemistry/methods , Microscopy, Confocal/methods , Muscle, Smooth/drug effects , Myosin Type II/analysis , Phorbol 12,13-Dibutyrate/pharmacology , Thapsigargin/pharmacology
7.
Can J Physiol Pharmacol ; 80(1): 1-7, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11911220

ABSTRACT

Selected parameters of cardiovascular function were evaluated in vitamin A-deficient rats at 70 days of age. Resting heart rate was increased by an average of 100 bpm (21.4+/-2.7%), whereas resting systolic blood pressure was normal in vitamin A-deficient animals. The maximal contractile force developed per milligram weight of tissue by aortic rings excised from vitamin A-deficient animals was reduced in response to high potassium (-25.0+/-8.7%) and phorbol 12,13-dibutyrate (-36.8+/-8.4%) but was only slightly reduced in response to norepinephrine (-17.8+/-11.1%). Intimal rubbing to remove the endothelium had no effect on the loss in contractile responsiveness, and the relaxant response to acetylcholine was similar between control and vitamin A-deficient tissue groups. This suggests that the decrease in contractility of vascular smooth muscle from the vitamin A-deficient rats did not involve altered release of endothelium-derived vasoactive factors. Western blot analysis suggested a reduction in the protein levels of several differentiation markers including alpha-actin (-22%), calponin (-37%), desmin (-37%), and vinculin (-40%), whereas the level of PKCalpha was unchanged from control values. Our findings indicate a significant decrease in contractile responsiveness of aortic smooth muscle of the vitamin A-deficient rat that may be associated with a down regulation in the expression of contractile-related proteins.


Subject(s)
Hemodynamics/physiology , Vitamin A Deficiency/physiopathology , Animals , Aorta, Thoracic/drug effects , Blood Pressure/physiology , Blotting, Western , Body Weight/physiology , Female , In Vitro Techniques , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Organ Size/physiology , Proteins/metabolism , Rats , Rats, Sprague-Dawley
8.
Acta Physiol Scand ; 174(3): 237-46, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11906323

ABSTRACT

Reports from numerous laboratories suggest that protein kinase C (PKC) translocation to substrate target sites may vary depending on cell type and experimental conditions. We have proposed that acutely variable targeting of PKC to different substrate sites could greatly expand the functional properties of individual isoforms in individual cell types (Li et al., 2001). Confocal microscopy and PKC alpha-enhanced green fluorescent protein (PKC alpha-EGFP) fusion protein expression were utilized to investigate the spatial and temporal pattern of PKC alpha translocation to different stimulating agents in A7r5 smooth muscle cells. Phorbol 12, 13 dibutyrate (PDBu 10(-8) M) caused a slow but irreversible relocation of the fusion protein from the cytosol to the plasmalemma. By comparison, thapsigargin (10(-5) M) and A23 187 (2 x 10(-5) M) induced a rapidly transient translocation to the cell membrane which was completed within 4 min. In contrast to these agents, angiotensin II (Ang II, 10(-6) M) caused only partial relocalization of cytosolic PKC alpha-EGFP to brightly fluorescing patches at the cell periphery. Localization at peripheral patches was completed within seconds and the fusion protein returned to the cytosol within 2 min. The PKC inhibitor staurosporine blocked cellular contraction to PDBu but not A(23 187) and had no effect on PKC alpha-EGFP translocation. By comparison, the calcium chelators EDTA and BAPTA-AM blocked the contraction to A(23 187), attenuated the contraction to PDBu, and abolished the translocation of PKC alpha-EGFP by both agents. The results show that in a single cell type the spatial and temporal characteristics of individual PKC isoform translocation may differ markedly. This further suggests the existence of potentially complex mechanisms which regulate the rate and location of target site availability.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Angiotensin II/pharmacology , Animals , Aorta/cytology , Aorta/embryology , Calcimycin/pharmacology , Carcinogens/pharmacology , Cell Line , Chelating Agents/pharmacology , Cytosol/drug effects , Cytosol/enzymology , Green Fluorescent Proteins , Isoenzymes/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Transport/drug effects , Rats , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Thapsigargin/pharmacology , Transfection
9.
Pflugers Arch ; 443(1): 38-47, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11692264

ABSTRACT

The subcellular translocation of PKCalpha was studied in A7r5 cells by confocal microscopy through use of standard immunohistologic staining and PKCalpha-enhanced green fluorescent protein (PKCalpha-EGFP) fusion protein expression. The results from both methods were consistent in indicating that PKCalpha, observed to be diffusely distributed in the unstimulated cell, was translocated primarily to either the perinuclear region of the cell or to subplasmalemmal sites depending on the concentration of phorbol 12, 13 dibutyrate (PDBu) used to activate the response. Translocation of PKCalpha to the perinucleus but not the plasmalemma was blocked by the use of colchicine to disrupt cell microtubules. However, there was little evidence of significant colocalization of PKCalpha with the microtubular cytoskeleton during the interval of translocation. By comparison, cytochalasin B disruption of actin microfilaments had no significant effect on PKCalpha translocation to either the plasmalemma or the perinucleus. The results indicate that the target site of PKCalpha translocation may vary with activating stimulus strength in A7r5 cells and that the translocation of the isoform to perinuclear target loci depends on an intact microtubular cytoskeleton. This suggests that multiple pathways are available for the redistribution of PKCalpha that may employ different mechanisms to regulate the movement and/or docking of the isoform at specific target sites.


Subject(s)
Isoenzymes/analysis , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/ultrastructure , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/analysis , Animals , Aorta , Cell Line , Cell Membrane/enzymology , Cell Nucleus/enzymology , Cytochalasin B/pharmacology , Cytoskeleton/enzymology , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Activation/drug effects , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Green Fluorescent Proteins , Isoenzymes/metabolism , Luminescent Proteins/genetics , Microscopy, Confocal , Protein Kinase C/metabolism , Rats , Recombinant Fusion Proteins , Transfection
10.
J Cell Physiol ; 189(2): 216-28, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11598907

ABSTRACT

The transcript (mRNA), protein levels, enzyme activity, and cellular localization of four protein kinase C (PKC) isozymes identified in rat osteogenic sarcoma cells (UMR-108) were studied at confluent density and during mechanical stress (cyclic stretch). Western blot analysis indicated that growth to confluent density significantly increased the protein levels of cPKC-alpha (11.6-fold), nPKC-delta (5.3-fold), and nPKC-epsilon (22.0-fold) but not aPKC-zeta. Northern blot analysis indicated a significant (2.3-fold) increase in the 10 kb transcript of cPKC-alpha, a slight (1.3-fold) increase in that of nPKC-epsilon but no detectable change in that of the remaining isozymes. Enzyme activity assays of the individually immunoprecipitated isozymes yielded detectable kinase activity only for PKC-alpha, PKC-delta, and PKC-epsilon and only in confluent cells, corroborating the selective increase of these isozymes at confluent density. The UMR-108 cells showed a dramatic orientation response to mechanical stress with cell reshaping and alignment of the cell long axis perpendicular to the axis of force, remodeling of the actin cytoskeleton, and the appearance of multiple peripheral sites which stained for actin, vinculin, and PKC in separate experiments. Longer term mechanical stress beyond 24 h, however, resulted in no significant change in the mRNA level, protein level, or enzyme activity of any of the four PKC isozymes investigated. The results indicate that there are isozyme-selective increases in the protein levels of PKC isozymes of osteoblastic UMR-108 cells upon growth to confluence which may be regulated at the transcriptional or the post-transcriptional level. The results from UMR-108 cells support the earlier proposal (Carvalho RS, Scott JE, Suga DM, Yen EH. 1994. J Bone Miner Res 9(7):999-1011) that PKC could be involved in the early phase of mechanotransduction in osteoblasts through the activation of focal adhesion assembly/disassembly and the remodeling of the actin cytoskeleton.


Subject(s)
Osteoblasts/enzymology , Protein Kinase C/biosynthesis , Protein Kinase C/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cell Culture Techniques/methods , Cell Size , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Microscopy, Confocal , Osteoblasts/cytology , Osteoblasts/ultrastructure , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Kinase C-delta , Protein Kinase C-epsilon , RNA, Messenger/biosynthesis , Rats , Stress, Mechanical , Transcription, Genetic , Tumor Cells, Cultured
11.
J Muscle Res Cell Motil ; 22(6): 521-34, 2001.
Article in English | MEDLINE | ID: mdl-12038586

ABSTRACT

Previous work has shown that stimulation of contraction in A7r5 smooth muscle cells with phorbol ester (PDBu) results in the disassembly and remodeling of the alpha-actin component of the cytoskeleton (Fultz et al., 2000, J Mus Res Cell Motil 21: 775-781). In the present study, we evaluated the effect of increasing intracellular calcium ion concentration [Ca2+]i by A23187 and thapsigargin on alpha- and beta-actin remodeling. The effects of A23187 and thapsigargin on cell contraction and actin remodeling were effectively identical. The two compounds caused contraction of A7r5 cells that was earlier in onset and more quickly completed than PDBu-induced contractions. Both the alpha- and beta-actin isoforms were incorporated into stress cables in the resting cell. During the interval of contraction, beta-actin cables shortened without evidence of disassembly. By comparison, the increase of [Ca2+]i resulted in partial or complete dissolution of alpha-actin cables without further remodeling. In addition, PDBu-mediated alpha-actin remodeling was blocked in the presence of A23187. Increased [Ca2+]i also caused dispersal of alpha-actinin but had no effect on the cellular distribution of talin suggesting the effect was selective for alpha-actin cytoskeletal structure. The incubation of cells in calcium-free media prevented alpha-actin dissolution by A23187/thapsigargin and also blocked PDBu-mediated remodeling. Finally, of six kinase inhibitors investigated, only ML-7 partially blocked the dissolution of alpha-actin cables by increased [Ca2+]i. The results suggest that the sustained elevation of [Ca2+]i beyond a threshold level initiates depolymerization of alpha-actin but not beta-actin. It further appears that PDBu-induced alpha-actin remodeling requires Ca2+ but increases of [Ca2+]i beyond a threshold level may inhibit this activity. The finding that ML-7 partially inhibits alpha-actin dissolution in the presence of A23187/thapsigargin may be suggesting that myosin light chain kinase (MLCK) plays a role in destabilizing alpha-actin structure in the activated cell.


Subject(s)
Actins/metabolism , Calcium/physiology , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Ionophores/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Thapsigargin/pharmacology
12.
Acta Physiol Scand ; 170(2): 87-97, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11114946

ABSTRACT

The translocation of protein kinase C (PKC) isozymes from their inactive cell locus to a variety of cytoskeletal, organelle, and plasmalemmal sites is thought to play an important role in their activation and substrate specificity. We have utilized confocal microscopy to compare phorbol 12, 13 dibutyrate (PDB) - stimulated translocation of PKCalpha in cultured cells derived from rat vascular smooth muscle. In enzymatically dispersed, passaged smooth muscle cells, PKCalpha was uniformly distributed throughout the unstimulated cell. PDB stimulation resulted in extensive association of the PKCalpha into filamentous strands with subsequent accumulation of the isoform in the peri-nuclear region of the cell. Dual immunostaining indicated that PKCalpha was extensively colocalized with microtubules in the interval immediately following PDB stimulation but was largely disassociated from microtubules at 10 min, at which time the translocation of PKCalpha to the peri-nucleus/nucleus was nearly complete. It was further found that the use of colchicine to disrupt the microtubules caused the loss of PKCalpha translocation to the peri-nuclear region. By comparison, cytochalasin B disruption of actin microfilaments had no significant effect on this parameter. The data suggest that PDB stimulation results in a transient association of PKCalpha with cell microtubules and that the microtubules play an important role in the translocation of PKCalpha from the cytosol in passaged cells derived from rat aortic smooth muscle.


Subject(s)
Isoenzymes/metabolism , Microtubules/metabolism , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Aorta/cytology , Biological Transport/physiology , Cells, Cultured , Colchicine/pharmacology , Cytochalasin B/pharmacology , Cytosol/metabolism , Male , Microtubules/drug effects , Muscle, Smooth, Vascular/cytology , Protein Kinase C-alpha , Rats , Rats, Sprague-Dawley
13.
Can J Physiol Pharmacol ; 78(6): 500-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10914640

ABSTRACT

The effect of short-term exposure to homocysteine (Hcy) on the contractile characteristics of rat aortic tissue was assessed both in vitro and in vivo. The contractile response of Hcy-treated aortic rings in culture for 1 or 4 days was unchanged from control responses. By comparison, aortic rings from animals injected with Hcy showed marked attenuation of response compared with controls injected with saline, cysteine or methionine. The contractile response to K+ was decreased within 24 hours of Hcy injection, whereas the response to both K+ (-27%) and noradrenaline (-56%) was significantly decreased by 4 days. In contrast, the contractile response to phorbol-12,13-dibutyrate was not different between Hcy and control groups. Intimal rubbing completely restored the responsiveness of Hcy-treated tissue to K+ and noradrenaline. By comparison, L-NAME only partially restored contractile responsiveness, while the cyclooxygenase inhibitor indomethacin had no effect on contractile attenuation induced by Hcy. Western blot analysis showed a 2-fold increase of endothelial nitric oxide synthase (eNOS) and a 3-fold increase in inducible nitric oxide synthase (iNOS) protein expression in the aortic endothelial cells from Hcy-injected rats. The results indicate an early detectable effect of Hcy on the in vivo contractile properties of vascular smooth muscle. The effect is endothelium-mediated and may vary depending on the agonist studied. The mechanism is uncertain but appears to involve increased nitric oxide (NO) production. Finally, the data suggest that attenuation of contraction may not be due to a direct effect of Hcy but that the compound is modified or acts indirectly in vivo.


Subject(s)
Aorta/drug effects , Endothelium, Vascular/drug effects , Homocysteine/pharmacology , Vasoconstriction/drug effects , Animals , Aorta/physiology , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Norepinephrine/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
14.
J Muscle Res Cell Motil ; 21(8): 775-87, 2000.
Article in English | MEDLINE | ID: mdl-11392559

ABSTRACT

It has been proposed that the reorganization of components of the actin cytomatrix could contribute to force development and the low energy cost of sustained contraction in contractile cells which lack a structured sarcomere (A.S. Battistella-Patterson, S. Wang and G.L. Wright (1997) Can J Physiol Pharmacol 75: 1287-1299). However, there has been no direct evidence of an apropos actin reorganization specifically linked to the contractile response in cells of this type. Remodeling of the alpha- and beta-actin domains was studied in A7r5 smooth muscle cells during phorbol 12,13 dibutyrate (PDB)-induced contraction using immunohistologic staining and beta-actin-green fluorescent protein (beta-actin-GFP) fusion protein expression. Cell stained with phalloidin as well as cells expressing beta-actin-GFP showed densely packed actin stress cables, arranged in parallel and extending across the cell body. PDB caused approximately 85% of cells to contract with evidence of forcible detachment from peripheral adhesion sites seen in many cells. The contraction of the cell body was not uniform but occurred along a principal axis parallel to the system of densely packed beta-actin cables. During the interval of contraction, the beta-actin cables shortened without evidence of disassembly or new cable formation. The use of cytochalasin to inhibit actin polymerization resulted in the dissolution of the actin cables at the central region of the cell and caused the elongation of precontracted cells. In unstimulated cells, alpha-actin formed cables similar in arrangement to the cell spanning beta-actin cables. Within a short interval after PDB addition; however, the majority of alpha-actin cables disassembled and reformed into intensely fluorescing column-like structures extending vertically from the cell base at the center of clusters of alpha-actin filaments. The alpha-actin columns of contracting cells showed strong colocalization of alpha-actinin suggesting they could be structurally analogous to the dense bodies of highly differentiated smooth muscle cells. The results indicate that the alpha- and beta-actin domains of A7r5 cells undergo a highly structured reorganization during PDB-induced contraction. The extent and nature of this restructuring suggest that remodeling could play a role in contractile function.


Subject(s)
Actins/physiology , Cytoskeleton/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Animals , Aorta , Carcinogens/pharmacology , Cell Size/drug effects , Cell Size/physiology , Cells, Cultured , Cytoskeleton/drug effects , Embryo, Mammalian , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...