Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Microsc ; 228(Pt 3): 406-12, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18045335

ABSTRACT

We report the successful implementation of a fully automated tomographic data collection system in scanning transmission electron microscopy (STEM) mode. Autotracking is carried out by combining mechanical and electronic corrections for specimen movement. Autofocusing is based on contrast difference of a focus series of a small sample area. The focus gradient that exists in normal images due to specimen tilt is effectively removed by using dynamic focusing. An advantage of STEM tomography with dynamic focusing over TEM tomography is its ability to reconstruct large objects with a potentially higher resolution.

2.
FEBS Lett ; 578(1-2): 73-9, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15581619

ABSTRACT

Ca2+-sensitization of smooth muscle occurs through inhibition of myosin light chain phosphatase (MLCP) leading to an increase in the MLCK:MLCP activity ratio. MLCP is inhibited through phosphorylation of its regulatory subunit (MYPT-1) following activation of the RhoA/Rho kinase (ROK) pathway or through phosphorylation of the PP1c inhibitory protein, CPI-17, by PKC delta or ROK. Here, we explore the crosstalk between these two modes of MLCP inhibition in a smooth muscle of a natural CPI-17 knockout, chicken amnion. GTPgammaS elicited Ca2+-sensitized force which was relaxed by GDI or Y-27632, however, U46619, carbachol and phorbol ester failed to induce Ca2+-sensitized force, but were rescued by recombinant CPI-17, and were sensitive to Y-27632 inhibition. In the presence, but not absence, of CPI-17, U46619 also significantly increased GTP.RhoA. There was no affect on MYPT-1 phosphorylation at T695, however, T850 phosphorylation increased in response to GTPgammaS stimulation. Together, these data suggest a role for CPI-17 upstream of RhoA activation possibly through activation of another PP1 family member targeted by CPI-17.


Subject(s)
Amnion/anatomy & histology , Calcium/metabolism , Cyclic GMP/analogs & derivatives , Muscle Proteins/metabolism , Muscle, Smooth/physiology , Phosphoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , rhoA GTP-Binding Protein/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Aluminum Compounds/pharmacology , Animals , Bacterial Toxins/metabolism , Chickens/anatomy & histology , Chickens/physiology , Cyclic GMP/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fluorides/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Triphosphate/metabolism , In Vitro Techniques , Muscle Contraction/physiology , Muscle Proteins/genetics , Muscle, Smooth/drug effects , Myosin-Light-Chain Phosphatase/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoproteins/genetics , Protein Phosphatase 1 , Protein Subunits/metabolism , Signal Transduction/physiology , Vasoconstrictor Agents/pharmacology
3.
Structure ; 12(11): 1955-65, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15530360

ABSTRACT

Calcium sensitization in smooth muscle is mediated by the RhoA GTPase, activated by hitherto unspecified nucleotide exchange factors (GEFs) acting downstream of Galphaq/Galpha(12/13) trimeric G proteins. Here, we show that at least one potential GEF, the PDZRhoGEF, is present in smooth muscle, and its isolated DH/PH fragment induces calcium sensitization in the absence of agonist-mediated signaling. In vitro, the fragment shows high selectivity for the RhoA GTPase. Full-length fragment is required for the nucleotide exchange, as the isolated DH domain enhances it only marginally. We crystallized the DH/PH fragment of PDZRhoGEF in complex with nonprenylated human RhoA and determined the structure at 2.5 A resolution. The refined molecular model reveals that the mutual disposition of the DH and PH domains is significantly different from other previously described complexes involving DH/PH tandems, and that the PH domain interacts with RhoA in a unique mode. The DH domain makes several specific interactions with RhoA residues not conserved among other Rho family members, suggesting the molecular basis for the observed specificity.


Subject(s)
Calcium/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Muscle, Smooth/physiology , rhoA GTP-Binding Protein/chemistry , Blotting, Western , Crystallography, X-Ray , Guanine Nucleotide Exchange Factors/chemistry , Humans , Models, Molecular , Protein Conformation , Rho Guanine Nucleotide Exchange Factors , rhoA GTP-Binding Protein/metabolism
4.
Biophys J ; 86(4): 2318-28, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15041670

ABSTRACT

The effects of myosin regulatory light chain (RLC) phosphorylation and strain on adenosine diphosphate (ADP) release from cross-bridges in phasic (rabbit bladder (Rbl)) and tonic (femoral artery (Rfa)) smooth muscle were determined by monitoring fluorescence transients of the novel ADP analog, 3'-deac-eda-ADP (deac-edaADP). Fluorescence transients reporting release of 3'-deac-eda-ADP were significantly faster in phasic (0.57 +/- 0.06 s(-1)) than tonic (0.29 +/- 0.03 s(-1)) smooth muscles. Thiophosphorylation of regulatory light chains increased and strain decreased the release rate approximately twofold. The calculated (k-ADP/k+ADP) dissociation constant, Kd of unstrained, unphosphorylated cross-bridges for ADP was 0.6 microM for rabbit bladder and 0.3 microM for femoral artery. The rates of ADP release from rigor bridges and reported values of Pi release (corresponding to the steady-state adenosine triphosphatase (ATPase) rate of actomyosin (AM)) from cross-bridges during a maintained isometric contraction are similar, indicating that the ADP-release step or an isomerization preceding it may be limiting the adenosine triphosphatase rate. We conclude that the strain- and dephosphorylation-dependent high affinity for and slow ADP release from smooth muscle myosin prolongs the fraction of the duty cycle occupied by strongly bound actomyosin.ADP state(s) and contributes to the high economy of force.


Subject(s)
Adenosine Diphosphate/metabolism , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Myosin Light Chains/metabolism , Smooth Muscle Myosins/metabolism , Actomyosin/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Fluorescent Dyes/chemistry , Muscle, Skeletal/metabolism , Phosphorylation , Rabbits , Urinary Bladder/metabolism
6.
J Muscle Res Cell Motil ; 25(8): 657-65, 2004.
Article in English | MEDLINE | ID: mdl-15750850

ABSTRACT

Telokin, a 17 kDa smooth muscle specific protein, consists of the C-terminal domain of MLCK, is phosphorylated by PKA and PKG at Ser13 in vivo (Wu et al. (1998) J Biol Chem 273: 11362-11369; Walker et al. (2001) J. Biol Chem 276: 24519-24524) and is proposed to induce Ca2+-desensitization through activation of myosin phosphatase (Wu et al. (1998) J. Biol Chem 273: 11362-11369). Telokin is reported to be highly expressed in phasic with only trace amounts in tonic smooth muscle. In alpha-toxin permeabilized femoral artery, 5 microM 8-Br-cGMP induced a two-fold increase in telokin phosphorylation and a maximal 30% relaxation of Ca2+-activated force compared to a 90% relaxation in phasic ileum muscle consistent with the relative amounts of telokin expressed in ileum, 27+/-4.6 microM SEM compared to 6+/-1.7 microM SEM, in femoral artery. Recombinant Wt telokin and the phospho-telokin mutant, S13D relaxed telokin-depleted femoral artery, by 38+/-8% SEM and 60+/-20% SEM, respectively. 8-Br-cGMP increased the rate and decreased the amplitude of force development initiated by photolysis of caged ATP in alpha-toxin permeabilized ileum and femoral artery smooth muscle, consistent with a cGMP-induced increase in phosphatase activity. Similarly, in telokin depleted ileum, recombinant S13D mutant telokin significantly increased the rate (0.08+/-0.01 s-1 vs. 014+/-0.02 s-1) and decreased force amplitude. In conclusion, our data support a role for telokin in cyclic nucleotide-induced relaxation of not only phasic, but also tonic smooth muscle and that this relaxation is mediated by activation of myosin phosphatase activity leading to a decrease in myosin light chain phosphorylation.


Subject(s)
Calcium/physiology , Muscle, Smooth/physiology , Myosin-Light-Chain Kinase/physiology , Myosin-Light-Chain Phosphatase/metabolism , Peptides/physiology , Adenosine Triphosphate/metabolism , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Enzyme Activation , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Kinase/biosynthesis , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Kinase/pharmacology , Peptide Fragments , Peptides/metabolism , Peptides/pharmacology , Phosphorylation , Rabbits , Recombinant Proteins/pharmacology , Time Factors
8.
Philos Trans R Soc Lond B Biol Sci ; 359(1452): 1921-30, 2004 Dec 29.
Article in English | MEDLINE | ID: mdl-15647168

ABSTRACT

The relationship of the biochemical states to the mechanical events in contraction of smooth muscle cross-bridges is reviewed. These studies use direct measurements of the kinetics of Pi and ADP release. The rate of release of Pi from thiophosphorylated cycling cross-bridges held isometric was biphasic with turnovers of 1.8 s-1 and 0.3 s-1, reflecting properties and forces directly acting on cross-bridges through mechanisms such as positive strain and inhibition by high-affinity MgADP binding. Fluorescent transients reporting release of an ADP analogue 3'-deac-edaADP were significantly faster in phasic than in tonic smooth muscles. Thiophosphorylation of myosin regulatory light chains (RLCs) increased and positive strain decreased the release rate around twofold. The rates of ADP release from rigor cross-bridges and the steady-state Pi release from cycling isometric cross-bridges are similar, indicating that the ADP-release step or an isomerization preceding it may limit the ATPase rate. Thus ADP release in phasic and tonic smooth muscles is a regulated step with strain- and dephosphorylation-dependence. High affinity of cross-bridges for ADP and slow ADP release prolong the fraction of the duty cycle occupied by strongly bound AM.ADP state(s) and contribute to the high economy of force that is characteristic of smooth muscle. RLC thiophosphorylation led to structural changes in smooth muscle cross-bridges consistent with our findings that thiophosphorylation and strain modulate product release.


Subject(s)
Models, Biological , Muscle Contraction/physiology , Muscle, Smooth/physiology , Myosins/physiology , Actins/metabolism , Adenosine Diphosphate/metabolism , Animals , Kinetics , Myosins/metabolism , Phosphates/metabolism , Phosphorylation , Protein Binding
9.
J Biol Chem ; 279(7): 5146-51, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14610087

ABSTRACT

The cGMP-dependent protein kinase (PKG) is the main mediator of nitric oxide-induced relaxation of smooth muscle. Although this pathway is well established, the cellular action of PKG, nitric oxide, and cGMP is complex and not fully understood. A cross-talk between the cGMP-PKG and other pathways (e.g. cAMP-protein kinase A) seems to exist. We have explored cGMP- and cAMP-dependent relaxation of smooth muscle using PKG-deficient mice (cGKI-/-). In intact ileum strips of wild type mice (cGKI+/+), 8-Br-cGMP inhibited the sustained phase of carbachol contractions by approximately 80%. The initial peak was less inhibited (approximately 30%). This relaxation was associated with a reduction in intracellular [Ca2+] and decreased Ca2+ sensitivity. Contractions of cGKI-/- ileum were not influenced by 8-Br-cGMP. EC50 for 8-Br-cGMP for PKG was estimated to be 10 nm. PKG-independent relaxation by 8-Br-cGMP had an EC50 of 10 microm. Relaxation by cAMP (approximately 50% at 100 microm), Ca2+ sensitivity of force, and force potentiation by GTPgammaS were similar in cGKI+/+ and cGKI-/- tissues. The results show that PKG is the main target for cGMP-induced relaxation in intestinal smooth muscle. cGMP desensitize the contractile system to Ca2+ via PKG. PKG-independent pathways are activated at 1000-fold higher cGMP concentrations. Relaxation by cAMP can occur independently of PKG. Long term deficiency of PKG does not lead to an apparent up-regulation of the cAMP-dependent pathways or changes in Ca2+ sensitivity.


Subject(s)
Calcium/chemistry , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Muscle, Smooth/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/metabolism , Animals , Calcium/metabolism , Carbachol/metabolism , Carbachol/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Muscles/metabolism , Nucleotides/chemistry , Time Factors
10.
Physiol Rev ; 83(4): 1325-58, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14506307

ABSTRACT

Ca2+ sensitivity of smooth muscle and nonmuscle myosin II reflects the ratio of activities of myosin light-chain kinase (MLCK) to myosin light-chain phosphatase (MLCP) and is a major, regulated determinant of numerous cellular processes. We conclude that the majority of phenotypes attributed to the monomeric G protein RhoA and mediated by its effector, Rho-kinase (ROK), reflect Ca2+ sensitization: inhibition of myosin II dephosphorylation in the presence of basal (Ca2+ dependent or independent) or increased MLCK activity. We outline the pathway from receptors through trimeric G proteins (Galphaq, Galpha12, Galpha13) to activation, by guanine nucleotide exchange factors (GEFs), from GDP. RhoA. GDI to GTP. RhoA and hence to ROK through a mechanism involving association of GEF, RhoA, and ROK in multimolecular complexes at the lipid cell membrane. Specific domains of GEFs interact with trimeric G proteins, and some GEFs are activated by Tyr kinases whose inhibition can inhibit Rho signaling. Inhibition of MLCP, directly by ROK or by phosphorylation of the phosphatase inhibitor CPI-17, increases phosphorylation of the myosin II regulatory light chain and thus the activity of smooth muscle and nonmuscle actomyosin ATPase and motility. We summarize relevant effects of p21-activated kinase, LIM-kinase, and focal adhesion kinase. Mechanisms of Ca2+ desensitization are outlined with emphasis on the antagonism between cGMP-activated kinase and the RhoA/ROK pathway. We suggest that the RhoA/ROK pathway is constitutively active in a number of organs under physiological conditions; its aberrations play major roles in several disease states, particularly impacting on Ca2+ sensitization of smooth muscle in hypertension and possibly asthma and on cancer neoangiogenesis and cancer progression. It is a potentially important therapeutic target and a subject for translational research.


Subject(s)
Calcium/metabolism , GTP-Binding Proteins/metabolism , Myosin Light Chains/metabolism , Nonmuscle Myosin Type IIA/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Animals , Humans , Muscle, Skeletal/metabolism , Muscle, Smooth/metabolism , Myosin-Light-Chain Phosphatase
11.
J Biol Chem ; 278(41): 39892-6, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12907680

ABSTRACT

The purpose of this study was to determine whether steric blockage of one head by the second head of native two-headed myosin was responsible for the inactivity of nonphosphorylated two-headed myosin compared with the high activity of single-headed myosin, as suggested on the basis of electron microscopy of two-dimensional crystals of heavy meromyosin (Wendt, T., Taylor, D., Messier, T., Trybus, K. M., and Taylor, K. A. (1999) J. Cell Biol. 147, 1385-1390; and Wendt, T., Taylor, D., Trybus, K. M., and Taylor, K. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 4361-4366). Our earlier cryo-atomic force microscopy (cryo-AFM) (Zhang, Y., Shao, Z., Somlyo, A. P., and Somlyo, A. V. (1997) Biophys. J. 72, 1308-1318) indicates that thiophosphorylation of the regulatory light chain increases the separation of the two heads of a single myosin molecule, but the thermodynamic probability of steric hindrance by strong binding between the two heads was not determined. We now report this probability determined by cryo-AFM of single whole myosin molecules shown to have normal low ATPase activity (0.007 s-1). We found that the thermodynamic probability of the relative head positions of nonphosphorylated myosin was approximately equal between separated heads as compared with closely apposed heads (energy difference of 0.24 kT (where k is a Boltzman constant and T is the absolute temperature)), and thiophosphorylation increased the number of molecules having separated heads (energy advantage of -1.2 kT (where k is a Boltzman constant and I is the absolute temperature)). Our results do not support the suggestion that strong binding of one head to the other stabilizes the blocked conformation against thermal fluctuations resulting in steric blockage that can account for the low activity of nonphosphorylated two-headed myosin.


Subject(s)
Smooth Muscle Myosins/chemistry , Smooth Muscle Myosins/ultrastructure , Animals , Freezing , In Vitro Techniques , Microscopy, Atomic Force/methods , Muscle, Smooth/metabolism , Phosphorylation , Protein Conformation , Smooth Muscle Myosins/metabolism , Thermodynamics , Turkeys
12.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 5): 876-80, 2003 May.
Article in English | MEDLINE | ID: mdl-12777804

ABSTRACT

Mutants of the small G protein RhoA that are deficient in GTPase activity and thereby exhibit constitutive molecular signaling activity are commonly used to discover its cellular functions. In particular, two such mutants, Gly14-->Val (G14V) and Gln63-->Leu (Q63L), are often used interchangeably for such studies. However, while their in vitro rates of GTP hydrolysis are very similar, differences are observed in their other functional properties. The structure of G14V-RhoA is known; in order to assess whether structural variations are responsible for functional differences, the crystal structure of a Q63L-RhoA bound to the GTP-analog 5'-guanylylimidodiphosphate (GMPPNP) was determined at 1.5 A resolution. Overall, the structure is very similar to that of G14V-RhoA, but the significantly higher resolution data permit an improved basis for structural analysis and comparison. The data support the notion that differences observed between the mutants in vivo are likely to arise from altered affinities for RhoGDI and not from direct structural differences.


Subject(s)
rhoA GTP-Binding Protein/chemistry , Amino Acid Substitution , Crystallography, X-Ray , Escherichia coli/metabolism , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Dissociation Inhibitors/biosynthesis , Guanine Nucleotide Dissociation Inhibitors/chemistry , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanylyl Imidodiphosphate/metabolism , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Yeasts/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
13.
Am J Physiol Cell Physiol ; 285(3): C674-85, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12760907

ABSTRACT

An 80-kDa protein, prominently expressed in smooth muscle, was microsequenced and identified as LPP, the product of the lipoma-preferred partner gene (Petit MMR, Mols R, Schoenmakers EFPM, Mandahl N, and Van de Ven WJM. Genomics 36: 118-129, 1996). Using a specific anti-LPP antibody, we showed, in Western blots and with immunofluorescence microscopy, the selective expression of LPP in vascular and visceral smooth muscles (approximately 0.5-1 ng/microg total protein). In other mature (noncultured) tissues, including heart and skeletal muscle, the protein is present only in trace amounts and is closely correlated with the levels of the smooth muscle marker alpha-actin. In freshly isolated guinea pig bladder smooth muscle cells, immunofluorescence images showed LPP as linear arrays of punctate, longitudinally oriented staining superimposed with vinculin staining on the plasma membrane surface. A corresponding pattern of periodic labeling at the membrane in transverse sections of bladder smooth muscle suggested an association of LPP with peripheral dense bodies. In cultured rat aortic smooth muscle cells, LPP colocalized with vinculin at focal adhesions but not with p120 catenin or alpha-actinin. Overexpression of the protein increased EGF-stimulated migration of vascular smooth muscle cells in Transwell assays, suggesting the participation of LPP in cell motility. The Rho-kinase inhibitor Y-27632 dissociated focal adhesions and LPP staining at the cell periphery and enhanced the nuclear accumulation of LPP induced by leptomycin B, indicating that LPP has a potential for relocating to the nucleus through a shuttling mechanism that is sensitive to inhibition of Rho-kinase.


Subject(s)
Cytoskeletal Proteins/genetics , Muscle, Smooth, Vascular/physiology , Urinary Bladder/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Aorta, Thoracic/cytology , Cell Movement/physiology , Cell Nucleus/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Fatty Acids, Unsaturated/pharmacology , Fluorescent Antibody Technique , Gene Expression , Guinea Pigs , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Male , Molecular Weight , Muscle, Smooth, Vascular/cytology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rabbits , Urinary Bladder/cytology , rho-Associated Kinases
14.
Circulation ; 107(13): 1810-5, 2003 Apr 08.
Article in English | MEDLINE | ID: mdl-12665494

ABSTRACT

BACKGROUND: Ion channel remodeling occurs during atrial fibrillation (AF); however, the extent of alteration in the subcellular distribution of elements (Na, K, Cl, Ca, Mg, P) is unknown. Electron probe microanalysis was used to determine the total (free+bound) in vivo subcellular concentration of these elements during AF. METHODS AND RESULTS: The left atrial appendage (LAA) was snap-frozen in situ after pacing (640 bpm) for 3 minutes (n=5 dogs), 30 minutes (n=3), or 48 hours (n=5). Dogs in sinus rhythm (n=3) served as controls. Whole-cell, cytosolic, and mitochondrial elemental concentrations were measured in cryosections. LAA effective refractory period (ERP) was measured before and after pacing. LAA ERP decreased significantly after 48 hours (116+/-3 to 88+/-10 ms, P=0.02). Whole-cell Cl increased by 9.0 mmol/L and 17 mmol/L after 3 and 30 minutes of pacing, respectively (P<0.0001), without a concomitant increase in Na. However, at 48 hours, whole-cell Na was reduced by 51% (P<0.01). Cytosolic Ca increased by 1.1 mmol/kg dry wt after 3 minutes (P<0.005), but mitochondrial Ca remained low and unchanged. Cell size measured in transverse cryosections increased after 3 minutes of pacing (75+/-5 to 109+/-11 microm2, P=0.007) but returned to baseline by 30 minutes (66+/-5 microm2). CONCLUSIONS: Intracellular Cl accumulation induced by rapid pacing is a novel finding and may play a role in AF pathogenesis by causing resting membrane depolarization and ERP reduction. There was no evidence of cellular or mitochondrial Ca overload despite the development of electrical remodeling and transient increase in cytoplasmic Ca.


Subject(s)
Atrial Fibrillation/metabolism , Chlorides/metabolism , Animals , Atrial Appendage/chemistry , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Calcium/analysis , Cardiac Pacing, Artificial , Chlorides/analysis , Cytosol/chemistry , Dogs , Electron Probe Microanalysis , Electrophysiology , Female , Magnesium/analysis , Mitochondria/chemistry , Myocytes, Cardiac/pathology , Neurotransmitter Agents/physiology , Phosphorus/analysis , Potassium/analysis , Sodium/analysis
15.
Cell Calcium ; 33(4): 247-56, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12618145

ABSTRACT

We quantitated subcellular elemental concentrations in stimulated and resting guinea pig myocardium to determine whether species-specific properties of guinea pigs or the subcellular localization of mitochondria accounted for reports of higher mitochondrial Ca in guinea pigs than in other species. Small papillary muscles or trabeculae isolated from guinea pig ventricles were stimulated to raise cytosolic [Ca(2+)](i) by two methods: (1). tetanizing by rapid pacing preparations in which Ca(2+) uptake by the sarcoplasmic reticulum was inhibited with cyclopiazonic acid or (2). freeze trapping paced muscles near-peak systole. Electron probe X-ray microanalysis showed no significant difference between the (low, approximately 0.4 mmol/kg dry weight) mitochondrial Ca content of stimulated guinea pig hearts, compared to mitochondria of other species, such as rat and hamsters, and the Ca contents of peripheral and central mitochondria were also not significantly different.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Electron Probe Microanalysis , Enzyme Inhibitors/pharmacology , Guinea Pigs , Indoles/pharmacology , Male , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Myocardium/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Organ Culture Techniques , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure , Species Specificity , Up-Regulation/drug effects , Up-Regulation/physiology
16.
FASEB J ; 17(2): 223-34, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12554701

ABSTRACT

The purpose of this study was to determine the effects of inhibitors of Rho kinase (ROK) and matrix metalloproteinases (MMPs) on angiogenesis and tumor growth and to evaluate ROK activity in human prostate cancer PC3 cells and endothelial cells (HUVECs). Vacuolation by endothelial cells and lumen formation, the earliest detectable stages of angiogenesis, were inhibited by the ROK inhibitor Wf-536. Combining Wf-536 with the MMP inhibitor Marimastat greatly enhanced in vitro inhibition of endothelial vacuolation, lumen and cord formation, and VEGF- and HGF-stimulated endothelial sprout formation from aorta. Inhibition of sprout formation by the two inhibitors was synergistic. Both agents inhibited migration of HUVECs. The regulatory subunit (MYPT1) of the myosin phosphatase was phosphorylated in PC3 cells and HUVECs, and phosphorylation of MYPT1 and the myosin regulatory light chain was reduced by Wf-536, providing direct evidence of ROK activity. Early treatment of immuno-incompetent mice bearing xenotransplants of PC3 cells with a combination of Wf-536 plus Marimastat with or without Paclitaxel, significantly inhibited tumor growth, prevented tumor growth escape after discontinuation of Paclitaxel, and increased survival.


Subject(s)
Enzyme Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors , Neovascularization, Pathologic/prevention & control , Prostatic Neoplasms/prevention & control , Protein Serine-Threonine Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Cell Division/drug effects , Cell Line , Drug Synergism , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, SCID , Microscopy, Fluorescence , Myosin-Light-Chain Phosphatase , Neoplasm Transplantation , Paclitaxel/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Rabbits , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , rho-Associated Kinases
17.
Novartis Found Symp ; 246: 258-68; discussion 268-71, 272-6, 2002.
Article in English | MEDLINE | ID: mdl-12164313

ABSTRACT

Structural and functional studies indicate the important role of the sarcoplasmic reticulum (SR) in excitation-contraction coupling in smooth and striated muscles, as well as a similar Ca2+ signalling function of the endoplasmic reticulum (ER) in non-muscle cells. Electron probe analysis directly established the SR/ER of smooth muscle as a sink and source of Ca2+, while immunoelectron and immunofluorescence microscopy showed both inositol-1,4,5-trisphosphate (InsP3) and ryanodine receptors localized to its membranes. Structural relationships, some yet to have fully determined functions, occur between the mitochondria and the SR, and the junctional SR and plasma membrane. Ca2+ is released by stimuli that generate InsP3 indicating the primary role of InsP3 receptors in Ca2+-release in smooth, although not in striated, muscle. Pathological mitochondrial Ca2+ uptake occurs at high [Ca2+]i similarly in both muscle and non-muscle cells. Based on newer evidence, earlier experimental results obtained with fluorescent Ca2+ indicators and related to phasic and tonic components of contraction can now be reinterpreted. Electron energy loss spectroscopy for high-resolution Ca2+ imaging and flash photolysis of caged agonists for exploration of the rapid kinetics of Ca2+ release from the SR are currently being explored.


Subject(s)
Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Sarcoplasmic Reticulum/physiology , Animals , Cell Membrane/physiology , Chick Embryo , Heart/physiology , Mammals , Muscle, Smooth/physiology , Sarcoplasmic Reticulum/ultrastructure
18.
J Biol Chem ; 277(34): 30614-21, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12060659

ABSTRACT

A Rho-kinase inhibitor increases corpus cavernosum (CC) pressure in an in vivo rat model (Chitaley, K., Wingard, C. J., Webb, R. C., Branam, H., Stopper, V. S., Lewis, R. W., and Mills, T. M. (2001) Nat. Med. 7, 119-122) suggesting that Rho-mediated Ca(2+) sensitization of CC smooth muscle maintains the flaccid (contracted) state. We directly demonstrate Ca(2+) sensitization of permeabilized rabbit and human CC and identify a highly expressed molecular component of this pathway. Ca(2+) sensitization of force induced by endothelin or GTPgammaS was significantly greater in CC than in rabbit ileum smooth muscle and was accompanied by a 17-fold higher RhoA content. Pull-down assays with the RhoA binding domain of mDia showed the high RhoA content of CC to be available for activation by GTPgammaS. Ca(2+) sensitization induced by endothelin, phenylephrine, or GTPgammaS was completely relaxed by the Rho kinase inhibitor Y-27632. Human and rabbit CC both express the phosphatase inhibitor CPI-17, the myosin phosphatase regulatory (MYPT-1) and catalytic (PP1delta) subunits, and two isoforms of Rho kinase. We suggest that high expression of RhoA contributes, through RhoA-mediated Ca(2+) sensitization, to the flaccid state of CC that can be reversed by a water-soluble, orally active Rho kinase inhibitor suitable for therapy of erectile dysfunction.


Subject(s)
Calcium/metabolism , Cyclic GMP/analogs & derivatives , Penile Erection/physiology , rhoA GTP-Binding Protein/physiology , Animals , Cyclic GMP/pharmacology , Endothelins/pharmacology , Erectile Dysfunction/drug therapy , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Male , Rabbits , rhoA GTP-Binding Protein/antagonists & inhibitors
19.
J Biol Chem ; 277(14): 12479-85, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11805108

ABSTRACT

Binding of the urokinase-type plasminogen activator (uPA) to its receptor activates diverse cell signaling pathways. How these signals are integrated so that cell physiology is altered remains unclear. In this study, we demonstrated that migration of MCF-7 breast cancer cells and HT-1080 fibrosarcoma cells on serum-coated surfaces is stimulated by agents that activate ERK, including uPA, epidermal growth factor, and constitutively active MEK1. The promigratory activity of these agents was entirely blocked not only by the MEK-specific antagonist PD098059, but also by antagonists of the Rho-Rho kinase pathway, including Y-27632 and dominant-negative RhoA (RhoA-N19). uPA did not significantly increase the level of GTP-bound RhoA, suggesting that the constitutive activity of the Rho-Rho kinase pathway may be sufficient to support ERK-stimulated cell migration. Paradoxically, Y-27632 and RhoA-N19 increased ERK phosphorylation in MCF-7 cells, providing further evidence that ERK activation alone does not promote cell migration when Rho kinase is antagonized. When MCF-7 cell migration was stimulated by ERK-independent processes such as expression of the beta(3) integrin subunit or changing the substratum to type I collagen, Y-27632 and RhoA-N19 failed to inhibit the response. This study supports a model in which the Ras-ERK and Rho-Rho kinase pathways cooperate to promote cell migration. Neutralizing either pathway is sufficient to block the response to agents that stimulate cell migration by activating ERK.


Subject(s)
Extracellular Signal-Regulated MAP Kinases , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Urokinase-Type Plasminogen Activator/metabolism , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Amides/pharmacology , Cell Line , Cell Movement , Collagen/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Guanosine Triphosphate/metabolism , Humans , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...