335 results on '"Walsh, MP"'
Search Results
102. Computational analysis of human adenovirus type 22 provides evidence for recombination among species D human adenoviruses in the penton base gene.
- Author
-
Robinson CM, Rajaiya J, Walsh MP, Seto D, Dyer DW, Jones MS, and Chodosh J
- Subjects
- Base Sequence, Cluster Analysis, Evolution, Molecular, Humans, Molecular Sequence Data, Phylogeny, Sequence Analysis, DNA, Sequence Homology, Adenoviruses, Human genetics, Capsid Proteins genetics, DNA, Viral genetics, Recombination, Genetic
- Abstract
Recombination in human adenoviruses (HAdV) may confer virulence upon an otherwise nonvirulent strain. The genome sequence of species D HAdV type 22 (HAdV-D22) revealed evidence for recombination with HAdV-D19 and HAdV-D37 within the capsid penton base gene. Bootscan analysis demonstrated that recombination sites within the penton base gene frame the coding sequences for the two external hypervariable loops in the protein. A similar pattern of recombination was evident within other HAdV-D types but not other HAdV species. Further study of recombination among HAdVs is needed to better predict possible recombination events among wild-type viruses and adenoviral gene therapy vectors.
- Published
- 2009
- Full Text
- View/download PDF
103. Ca2+-independent contraction of longitudinal ileal smooth muscle is potentiated by a zipper-interacting protein kinase pseudosubstrate peptide.
- Author
-
Ihara E, Moffat L, Borman MA, Amon JE, Walsh MP, and MacDonald JA
- Subjects
- Animals, Apoptosis Regulatory Proteins chemistry, Apoptosis Regulatory Proteins genetics, Calcium metabolism, Calcium-Calmodulin-Dependent Protein Kinases chemistry, Calcium-Calmodulin-Dependent Protein Kinases genetics, Catalytic Domain, Chickens, Death-Associated Protein Kinases, Enzyme Activation, Ileum enzymology, In Vitro Techniques, Microcystins pharmacology, Muscle Proteins metabolism, Muscle, Smooth drug effects, Mutation, Myosin Light Chains metabolism, Myosin-Light-Chain Kinase pharmacology, Phosphoprotein Phosphatases metabolism, Phosphoproteins metabolism, Phosphorylation, Protein Conformation, Protein Kinase Inhibitors pharmacology, Protein Phosphatase 1 metabolism, Rats, Substrate Specificity, Apoptosis Regulatory Proteins metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Gastrointestinal Motility drug effects, Ileum drug effects, Muscle Contraction drug effects, Muscle, Smooth enzymology, Myosin-Light-Chain Kinase metabolism, Peptide Fragments pharmacology, Signal Transduction drug effects
- Abstract
As a regulator of smooth muscle contraction, zipper-interacting protein kinase (ZIPK) can directly phosphorylate the myosin regulatory light chains (LC20) and produce contractile force. Synthetic peptides (SM-1 and AV25) derived from the autoinhibitory region of smooth muscle myosin light chain kinase can inhibit ZIPK activity in vitro. Paradoxically, treatment of Triton-skinned ileal smooth muscle strips with AV25, but not SM-1, potentiated Ca2+-independent, microcystin- and ZIPK-induced contractions. The AV25-induced potentiation was limited to ileal and colonic smooth muscles and was not observed in rat caudal artery. Thus the potentiation of Ca2+-independent contractions by AV25 appeared to be mediated by a mechanism unique to intestinal smooth muscle. AV25 treatment elicited increased phosphorylation of LC20 (both Ser-19 and Thr-18) and myosin phosphatase-targeting subunit (MYPT1, inhibitory Thr-697 site), suggesting involvement of a Ca2+-independent LC20 kinase with coincident inhibition of myosin phosphatase. The phosphorylation of the inhibitor of myosin phosphatase, CPI-17, was not affected. The AV25-induced potentiation was abolished by pretreatment with staurosporine, a broad-specificity kinase inhibitor, but specific inhibitors of Rho-associated kinase, PKC, and MAPK pathways had no effect. When a dominant-negative ZIPK [kinase-dead ZIPK((1-320))-D161A] was added to skinned ileal smooth muscle, the potentiation of microcystin-induced contraction by AV25 was blocked. Furthermore, pretreatment of skinned ileal muscle with SM-1 abolished AV25-induced potentiation. We conclude, therefore, that, even though AV25 is an in vitro inhibitor of ZIPK, activation of the ZIPK pathway occurs following application of AV25 to permeabilized ileal smooth muscle. Finally, we propose a mechanism whereby conformational changes in the pseudosubstrate region of ZIPK permit augmentation of ZIPK activity toward LC(20) and MYPT1 in situ. AV25 or molecules based on its structure could be used in therapeutic situations to induce contractility in diseases of the gastrointestinal tract associated with hypomotility.
- Published
- 2009
- Full Text
- View/download PDF
104. Characterization of tightly associated smooth muscle myosin-myosin light-chain kinase-calmodulin complexes.
- Author
-
Hong F, Haldeman BD, John OA, Brewer PD, Wu YY, Ni S, Wilson DP, Walsh MP, Baker JE, and Cremo CR
- Subjects
- Actins metabolism, Adenosine Triphosphate pharmacology, Animals, Cattle, Chickens, Enzyme-Linked Immunosorbent Assay, Humans, Kinetics, Magnesium pharmacology, Myosin Light Chains metabolism, Myosin-Light-Chain Kinase chemistry, Myosin-Light-Chain Kinase pharmacology, Peptide Fragments pharmacology, Phosphorylation drug effects, Protein Binding drug effects, Protein Structure, Tertiary, Calmodulin metabolism, Multiprotein Complexes metabolism, Myosin-Light-Chain Kinase metabolism, Smooth Muscle Myosins metabolism
- Abstract
A current popular model to explain phosphorylation of smooth muscle myosin (SMM) by myosin light-chain kinase (MLCK) proposes that MLCK is bound tightly to actin but weakly to SMM. We found that MLCK and calmodulin (CaM) co-purify with unphosphorylated SMM from chicken gizzard, suggesting that they are tightly bound. Although the MLCK:SMM molar ratio in SMM preparations was well below stoichiometric (1:73+/-9), the ratio was approximately 23-37% of that in gizzard tissue. Fifteen to 30% of MLCK was associated with CaM at approximately 1 nM free [Ca(2+)]. There were two MLCK pools that bound unphosphorylated SMM with K(d) approximately 10 and 0.2 microM and phosphorylated SMM with K(d) approximately 20 and 0.2 microM. Using an in vitro motility assay to measure actin sliding velocities, we showed that the co-purifying MLCK-CaM was activated by Ca(2+) and phosphorylation of SMM occurred at a pCa(50) of 6.1 and at a Hill coefficient of 0.9. Similar properties were observed from reconstituted MLCK-CaM-SMM. Using motility assays, co-sedimentation assays, and on-coverslip enzyme-linked immunosorbent assays to quantify proteins on the motility assay coverslip, we provide strong evidence that most of the MLCK is bound directly to SMM through the telokin domain and some may also be bound to both SMM and to co-purifying actin through the N-terminal actin-binding domain. These results suggest that this MLCK may play a role in the initiation of contraction.
- Published
- 2009
- Full Text
- View/download PDF
105. Genomic and bioinformatics analyses of HAdV-14p, reference strain of a re-emerging respiratory pathogen and analysis of B1/B2.
- Author
-
Seto J, Walsh MP, Mahadevan P, Purkayastha A, Clark JM, Tibbetts C, and Seto D
- Subjects
- Adenoviruses, Human pathogenicity, Base Sequence, Capsid Proteins analysis, Capsid Proteins genetics, DNA, Viral analysis, DNA, Viral genetics, Genome, Viral, Humans, Inverted Repeat Sequences, Molecular Sequence Data, Phylogeny, Proteome analysis, Sequence Alignment, Sequence Homology, Nucleic Acid, Virulence, Adenovirus Infections, Human virology, Adenoviruses, Human genetics, Computational Biology, Genomics
- Abstract
Unlike other human adenovirus (HAdV) species, B is divided into subspecies B1 and B2. Originally this was partly based on restriction enzyme (RE) analysis. B1 members, except HAdV-50, are commonly associated with respiratory diseases while B2 members are rarely associated with reported respiratory diseases. Recently two members of B2 have been identified in outbreaks of acute respiratory disease (ARD). One, HAdV-14, has re-emerged after an apparent 52-year absence. Genomic analysis and bioinformatics data are reported for HAdV-14 prototype for use as a reference and to understand and counter its re-emergence. The data complement and extend the original criteria for subspecies designation, unique amongst the adenoviruses, and highlight differences between B1 and B2, representing the first comprehensive analysis of this division. These data also provide finer granularity into the pathoepidemiology of the HAdVs. Whole genome analysis uncovers heterogeneous identity structures of the hexon and fiber genes amongst the HAdV-14 and the B1/B2 subspecies, which may be important in prescient vaccine development. Analysis of cell surface proteins provides insight into HAdV-14 tropism, accounting for its role as a respiratory pathogen. This HAdV-14 prototype genome is also a reference for applications of B2 adenoviruses as vectors for vaccine development and gene therapy.
- Published
- 2009
- Full Text
- View/download PDF
106. Identification and functional characterization of protein kinase A-catalyzed phosphorylation of potassium channel Kv1.2 at serine 449.
- Author
-
Johnson RP, El-Yazbi AF, Hughes MF, Schriemer DC, Walsh EJ, Walsh MP, and Cole WC
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Humans, Kidney cytology, Kv1.2 Potassium Channel chemistry, Membrane Potentials physiology, Molecular Sequence Data, Mutagenesis, Site-Directed, Patch-Clamp Techniques, Phosphorylation physiology, Protein Structure, Tertiary, Rabbits, Serine metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Kv1.2 Potassium Channel genetics, Kv1.2 Potassium Channel metabolism
- Abstract
Vascular smooth muscle Kv1 delayed rectifier K+ channels (KDR) containing Kv1.2 control membrane potential and thereby regulate contractility. Vasodilatory agonists acting via protein kinase A (PKA) enhance vascule smooth muscle Kv1 activity, but the molecular basis of this regulation is uncertain. We characterized the role of a C-terminal phosphorylation site, Ser-449, in Kv1.2 expressed in HEK 293 cells by biochemical and electrophysiological methods. We found that 1) in vitro phosphorylation of Kv1.2 occurred exclusively at serine residues, 2) one major phosphopeptide that co-migrated with 449pSASTISK was generated by proteolysis of in vitro phosphorylated Kv1.2, 3) the peptide 445KKSRSASTISK exhibited stoichiometric phosphorylation by PKA in vitro, 4) matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectroscopy (MS) and MS/MS confirmed in vitro Ser-449 phosphorylation by PKA, 5) in situ phosphorylation at Ser-449 was detected in HEK 293 cells by MALDI-TOF MS followed by MS/MS. MIDAS (multiple reaction monitoring-initiated detection and sequencing) analysis revealed additional phosphorylated residues, Ser-440 and Ser-441, 6) in vitro 32P incorporation was significantly reduced in Kv1.2-S449A, Kv1.2-S449D, and Kv1.2-S440A/S441A/S449A mutant channels, but Kv1.2-S440A/S441A was identical to wild-type Kv1.2 (Kv1.2-WT), and 7) bath applied 8-Br-cAMP or dialysis with PKA catalytic subunit (cPKA) increased Kv1.2-WT but not Kv1.2-S449A current amplitude. cPKA increased Kv1.2-WT current in inside-out patches. Rp-CPT-cAMPS reduced Kv1.2-WT current, blocked the increase due to 8-Br-cAMP, but had no effect on Kv1.2-S449A. cPKA increased current due to double mutant Kv1.2-S440A/S441A but had no effect on Kv1.2-S449D or Kv1.2-S440A/S441A/S449A. We conclude that Ser-449 in Kv1.2 is a site of PKA phosphorylation and a potential molecular mechanism for Kv1-containing KDR channel modulation by agonists via PKA activation.
- Published
- 2009
- Full Text
- View/download PDF
107. Evidence of molecular evolution driven by recombination events influencing tropism in a novel human adenovirus that causes epidemic keratoconjunctivitis.
- Author
-
Walsh MP, Chintakuntlawar A, Robinson CM, Madisch I, Harrach B, Hudson NR, Schnurr D, Heim A, Chodosh J, Seto D, and Jones MS
- Subjects
- Adenoviruses, Human genetics, Animals, Base Sequence, Computational Biology methods, Evolution, Molecular, Genome, Viral, Humans, Mice, Mice, Inbred C57BL, Models, Genetic, Molecular Sequence Data, Phylogeny, Sequence Homology, Nucleic Acid, Adenoviruses, Human metabolism, Keratoconjunctivitis etiology, Keratoconjunctivitis virology, Recombination, Genetic
- Abstract
In 2005, a human adenovirus strain (formerly known as HAdV-D22/H8 but renamed here HAdV-D53) was isolated from an outbreak of epidemic keratoconjunctititis (EKC), a disease that is usually caused by HAdV-D8, -D19, or -D37, not HAdV-D22. To date, a complete change of tropism compared to the prototype has never been observed, although apparent recombinant strains of other viruses from species Human adenovirus D (HAdV-D) have been described. The complete genome of HAdV-D53 was sequenced to elucidate recombination events that lead to the emergence of a viable and highly virulent virus with a modified tropism. Bioinformatic and phylogenetic analyses of this genome demonstrate that this adenovirus is a recombinant of HAdV-D8 (including the fiber gene encoding the primary cellular receptor binding site), HAdV-D22, (the epsilon determinant of the hexon gene), HAdV-D37 (including the penton base gene encoding the secondary cellular receptor binding site), and at least one unknown or unsequenced HAdV-D strain. Bootscanning analysis of the complete genomic sequence of this novel adenovirus, which we have re-named HAdV-D53, indicated at least five recombination events between the aforementioned adenoviruses. Intrahexon recombination sites perfectly framed the epsilon neutralization determinant that was almost identical to the HAdV-D22 prototype. Additional bootscan analysis of all HAdV-D hexon genes revealed recombinations in identical locations in several other adenoviruses. In addition, HAdV-D53 but not HAdV-D22 induced corneal inflammation in a mouse model. Serological analysis confirmed previous results and demonstrated that HAdV-D53 has a neutralization profile representative of the epsilon determinant of its hexon (HAdV-D22) and the fiber (HAdV-D8) proteins. Our recombinant hexon sequence is almost identical to the hexon sequences of the HAdV-D strain causing EKC outbreaks in Japan, suggesting that HAdV-D53 is pandemic as an emerging EKC agent. This documents the first genomic, bioinformatic, and biological descriptions of the molecular evolution events engendering an emerging pathogenic adenovirus.
- Published
- 2009
- Full Text
- View/download PDF
108. Ca2+ sensitization via phosphorylation of myosin phosphatase targeting subunit at threonine-855 by Rho kinase contributes to the arterial myogenic response.
- Author
-
Johnson RP, El-Yazbi AF, Takeya K, Walsh EJ, Walsh MP, and Cole WC
- Subjects
- Animals, Blood Pressure, Blotting, Western, Dose-Response Relationship, Drug, Male, Middle Cerebral Artery drug effects, Muscle Proteins metabolism, Myosin Light Chains metabolism, Phosphoproteins metabolism, Phosphorylation, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Protein Kinase Inhibitors pharmacology, Rats, Rats, Sprague-Dawley, Threonine, rho-Associated Kinases antagonists & inhibitors, Calcium Signaling drug effects, Middle Cerebral Artery enzymology, Protein Phosphatase 1 metabolism, Vasoconstriction drug effects, rho-Associated Kinases metabolism
- Abstract
Ca(2+) sensitization has been postulated to contribute to the myogenic contraction of resistance arteries evoked by elevation of transmural pressure. However, the biochemical evidence of pressure-induced increases in phosphorylated myosin light chain phosphatase (MLCP) targeting subunit 1 (MYPT1) and/or 17 kDa protein kinase C (PKC)-potentiated protein phosphatase 1 inhibitor protein (CPI-17) required to sustain this view is not currently available. Here, we determined whether Ca(2+) sensitization pathways involving Rho kinase (ROK)- and PKC-dependent phosphorylation of MYPT1 and CPI-17, respectively, contribute to the myogenic response of rat middle cerebral arteries. ROK inhibitors (Y27632, 0.03-10 micromol l(-1); H1152, 0.001-0.3 micromol l(-1)) and PKC inhibitors (GF109203X, 3 micromol l(-1); Gö6976; 10 micromol l(-1)) suppressed myogenic vasoconstriction between 40 and 120 mmHg. An improved, highly sensitive 3-step Western blot method was developed for detection and quantification of MYPT1 and CPI-17 phosphorylation. Increasing pressure from 10 to 60 or 100 mmHg significantly increased phosphorylation of MYPT1 at threonine-855 (T855) and myosin light chain (LC(20)). Phosphorylation of MYPT1 at threonine-697 (T697) and CPI-17 were not affected by pressure. Pressure-evoked elevations in MYPT1-T855 and LC(20) phosphorylation were reduced by H1152, but MYPT1-T697 phosphorylation was unaffected. Inhibition of PKC with GF109203X did not affect MYPT1 or LC(20) phosphorylation at 100 mmHg. Our findings provide the first direct, biochemical evidence that a Ca(2+) sensitization pathway involving ROK-dependent phosphorylation of MYPT1 at T855 (but not T697) and subsequent augmentation of LC(20) phosphorylation contributes to myogenic control of arterial diameter in the cerebral vasculature. In contrast, suppression of the myogenic response by PKC inhibitors cannot be attributed to block of Ca(2+) sensitization mediated by CPI-17 or MYPT1 phosphorylation.
- Published
- 2009
- Full Text
- View/download PDF
109. Rho-kinase-mediated suppression of KDR current in cerebral arteries requires an intact actin cytoskeleton.
- Author
-
Luykenaar KD, El-Rahman RA, Walsh MP, and Welsh DG
- Subjects
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine analogs & derivatives, 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine pharmacology, Actins antagonists & inhibitors, Actins drug effects, Amides pharmacology, Animals, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cerebral Arteries cytology, Cytochalasin D pharmacology, Delayed Rectifier Potassium Channels antagonists & inhibitors, Delayed Rectifier Potassium Channels drug effects, Enzyme Inhibitors pharmacology, Female, Nucleic Acid Synthesis Inhibitors pharmacology, Patch-Clamp Techniques, Pyridines pharmacology, Rats, Rats, Sprague-Dawley, Thiazolidines pharmacology, Uridine Triphosphate pharmacology, Vasoconstriction physiology, rho-Associated Kinases antagonists & inhibitors, rho-Associated Kinases drug effects, Actins metabolism, Cerebral Arteries metabolism, Cytoskeleton metabolism, Delayed Rectifier Potassium Channels metabolism, rho-Associated Kinases metabolism
- Abstract
This study examined the role of the actin cytoskeleton in Rho-kinase-mediated suppression of the delayed-rectifier K(+) (K(DR)) current in cerebral arteries. Myocytes from rat cerebral arteries were enzymatically isolated, and whole cell K(DR) currents were monitored using conventional patch-clamp electrophysiology. At +40 mV, the K(DR) current averaged 19.8 +/- 1.6 pA/pF (mean +/- SE) and was potently inhibited by UTP (3 x 10(-5) M). This suppression was observed to depend on Rho signaling and was abolished by the Rho-kinase inhibitors H-1152 (3 x 10(-7) M) and Y-27632 (3 x 10(-5) M). Rho-kinase was also found to concomitantly facilitate actin polymerization in response to UTP. We therefore examined whether actin dynamics played a role in the ability of Rho-kinase to suppress K(DR) current and found that actin disruption using either cytochalasin D (1 x 10(-5) M) or latrunculin A (1 x 10(-8) M) prevented current modulation. Consistent with our electrophysiological observations, both Rho-kinase inhibition and actin disruption significantly attenuated UTP-induced depolarization and constriction of cerebral arteries. We propose that UTP initiates Rho-kinase-mediated remodeling of the actin cytoskeleton and consequently suppresses the K(DR) current, thereby facilitating the depolarization and constriction of cerebral arteries.
- Published
- 2009
- Full Text
- View/download PDF
110. Radiofrequency ablation induces antigen-presenting cell infiltration and amplification of weak tumor-induced immunity.
- Author
-
Dromi SA, Walsh MP, Herby S, Traughber B, Xie J, Sharma KV, Sekhar KP, Luk A, Liewehr DJ, Dreher MR, Fry TJ, and Wood BJ
- Subjects
- Animals, Catheter Ablation, Cell Line, Tumor, Humans, Immunity, Innate, Mice, Neoplasm Metastasis pathology, Urinary Bladder Neoplasms pathology, Antigen Presentation immunology, Antigen-Presenting Cells immunology, Neoplasm Metastasis immunology, Neoplasm Metastasis prevention & control, Urinary Bladder Neoplasms immunology, Urinary Bladder Neoplasms surgery
- Abstract
Purpose: To evaluate the influence of subtotal radiofrequency (RF) ablation on a tumor-specific immune response in a murine tumor model and to explore the role of intratumoral dendritic cells (ITDCs) in mediating this effect., Materials and Methods: Animal work was performed according to an approved protocol and in compliance with the National Cancer Institute Animal Care and Use Committee guidelines and regulations. A murine urothelial carcinoma (MB49) model expressing the male minor histocompatibility (HY) antigen was inoculated subcutaneously in female mice. Fourteen days later, splenic T cells were analyzed with enzyme-linked immunosorbent spot for HY immune response (n = 57). In subsequent experiments, mice were randomized into control (n = 7), RF ablation, ITDC (n = 9), and RF ablation + ITDC (n = 9) groups and monitored for tumor growth. Eleven days after treatment, tumors were harvested for histologic and immunohistochemical analysis. Animals demonstrating complete tumor regression were rechallenged in the contralateral flank., Results: Animals treated with subtotal RF ablation showed significant increases in tumor-specific class I and II responses to HY antigens and tumor regression. RF ablation, ITDC, and combined groups demonstrated similar levels of antigen-presenting cell infiltration; all groups demonstrated greater levels of infiltration compared with untreated controls. ITDC injection also resulted in tumor regression. However, combination therapy did not enhance tumor regression when compared with either treatment alone. Rechallenged mice in RF ablation, ITDC, and combination groups demonstrated significant tumor growth inhibition compared with controls., Conclusion: Subtotal RF ablation treatment results in enhanced systemic antitumor T-cell immune responses and tumor regression that is associated with increased dendritic cell infiltration. ITDC injection mimics the RF ablation effect but does not increase immune responses when injected immediately after RF ablation.
- Published
- 2009
- Full Text
- View/download PDF
111. Myosin light chains are not a physiological substrate of AMPK in the control of cell structure changes.
- Author
-
Bultot L, Horman S, Neumann D, Walsh MP, Hue L, and Rider MH
- Subjects
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine analogs & derivatives, 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine pharmacology, AMP-Activated Protein Kinase Kinases, Animals, Cell Line, Dogs, Humans, Phosphorylation, Protein Kinase Inhibitors pharmacology, Protein Kinases genetics, Rats, Substrate Specificity, rho-Associated Kinases antagonists & inhibitors, rho-Associated Kinases metabolism, Myosin Light Chains metabolism, Protein Kinases metabolism
- Abstract
The kinetics of myosin regulatory light chain (MLC) phosphorylation by recombinant AMP-activated protein kinase (AMPK) were compared with commercial AMPK from rat liver and smooth muscle myosin light chain kinase (smMLCK). With identical amounts of activity units, initial rates of phosphorylation of MLC were at least 100-fold less with recombinant AMPK compared to smMLCK, whereas with rat liver AMPK significant phosphorylation was seen. In Madin-Darby Canine Kidney cells, AMPK activation led to an increase in MLC phosphorylation, which was decreased by a Rho kinase inhibitor without affecting AMPK activation. Therefore, MLC phosphorylation during energy deprivation does not result from direct phosphorylation by AMPK.
- Published
- 2009
- Full Text
- View/download PDF
112. Inhibition of rat aortic smooth muscle contraction by 2-methoxyestradiol.
- Author
-
Gui Y, Zheng XL, Zheng J, and Walsh MP
- Subjects
- 2-Methoxyestradiol, Animals, Aorta metabolism, Endothelium, Vascular enzymology, Enzyme Inhibitors pharmacology, Estradiol metabolism, Female, In Vitro Techniques, Male, Muscle, Smooth, Vascular drug effects, Myosin Light Chains metabolism, Nitric Oxide metabolism, Phenylephrine pharmacology, Phosphorylation, Protein Biosynthesis, Rats, Rats, Sprague-Dawley, Receptors, Estrogen metabolism, Signal Transduction, Transcription, Genetic, Tubulin Modulators pharmacology, Vasoconstrictor Agents pharmacology, Estradiol analogs & derivatives, Muscle, Smooth, Vascular metabolism, Vasoconstriction drug effects
- Abstract
Recent studies suggest that 2-methoxyestradiol (2-ME), an estrogen metabolite, has a similar inhibitory effect as 17beta-estradiol (E2) on vascular tone. However, it is not known whether 2-ME mediates the effects of E2 or by what mechanism 2-ME regulates smooth muscle contraction. Therefore, we compared the effects of 2-ME and E2 on rat aortic smooth muscle contraction. A preincubation with 2-ME (10 microM) for 1 h inhibited phenylephrine (PE)-induced tension in endothelium-intact, but not -denuded, tissues, whereas E2 inhibited PE-induced contraction in both preparations. The effects of 2-ME and E2 on endothelium-intact preparations were prevented by L-NAME hydrochloride (a nitric oxide synthase inhibitor). The 2-ME treatment reduced PE-induced phosphorylation of the 20-kDa myosin regulatory light chain. The inhibitory effects of 2-ME and E2 were not affected by ICI-182780 (an estrogen receptor antagonist) or actinomycin D (a gene transcription inhibitor); however, the effect of 2-ME, but not E2, was prevented by cycloheximide (a protein synthesis inhibitor). Furthermore, the effect of E2 was not blocked by 1-aminobenzotriazole (a cytochrome P-450 inhibitor) or Ro 41-0960 (a catechol-O-methyltransferase inhibitor). The effect of 2-ME was not mimicked by microtubule-interfering agents (nocodazole or Taxol). We conclude that 2-ME inhibits smooth muscle contractility through an endothelium- and nitric oxide-dependent mechanism, which does not involve estrogen receptors or microtubule disruption. The effect of 2-ME, but not E2, involves de novo protein synthesis. 2-ME does not mediate the inhibitory effect of E2 on smooth muscle contraction. These results support a potentially important role of 2-ME in the regulation of smooth muscle tone in the vasculature.
- Published
- 2008
- Full Text
- View/download PDF
113. AMP-activated protein kinase phosphorylates and desensitizes smooth muscle myosin light chain kinase.
- Author
-
Horman S, Morel N, Vertommen D, Hussain N, Neumann D, Beauloye C, El Najjar N, Forcet C, Viollet B, Walsh MP, Hue L, and Rider MH
- Subjects
- AMP-Activated Protein Kinases, Adenosine Triphosphate chemistry, Adenosine Triphosphate genetics, Adenosine Triphosphate metabolism, Animals, Aorta chemistry, Benzimidazoles pharmacology, Calcium metabolism, Calcium-Calmodulin-Dependent Protein Kinases antagonists & inhibitors, Calcium-Calmodulin-Dependent Protein Kinases chemistry, Calcium-Calmodulin-Dependent Protein Kinases genetics, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Calmodulin genetics, Calmodulin metabolism, Cattle, Cells, Cultured, Chickens, Male, Mice, Mice, Knockout, Multienzyme Complexes chemistry, Multienzyme Complexes genetics, Muscle Contraction drug effects, Muscle Tonus drug effects, Muscle Tonus physiology, Muscle, Smooth chemistry, Myocytes, Smooth Muscle chemistry, Myosin-Light-Chain Kinase chemistry, Myosin-Light-Chain Kinase genetics, Naphthalimides pharmacology, Phenylephrine pharmacology, Phosphorylation drug effects, Potassium Chloride pharmacology, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Rats, Rats, Wistar, Vasoconstriction drug effects, Vasoconstriction physiology, Vasoconstrictor Agents pharmacology, Aorta enzymology, Multienzyme Complexes metabolism, Muscle Contraction physiology, Muscle, Smooth enzymology, Myocytes, Smooth Muscle enzymology, Myosin-Light-Chain Kinase metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Smooth muscle contraction is initiated by a rise in intracellular calcium, leading to activation of smooth muscle myosin light chain kinase (MLCK) via calcium/calmodulin (CaM). Activated MLCK then phosphorylates the regulatory myosin light chains, triggering cross-bridge cycling and contraction. Here, we show that MLCK is a substrate of AMP-activated protein kinase (AMPK). The phosphorylation site in chicken MLCK was identified by mass spectrometry to be located in the CaM-binding domain at Ser(815). Phosphorylation by AMPK desensitized MLCK by increasing the concentration of CaM required for half-maximal activation. In primary cultures of rat aortic smooth muscle cells, vasoconstrictors activated AMPK in a calcium-dependent manner via CaM-dependent protein kinase kinase-beta, a known upstream kinase of AMPK. Indeed, vasoconstrictor-induced AMPK activation was abrogated by the STO-609 CaM-dependent protein kinase kinase-beta inhibitor. Myosin light chain phosphorylation was increased under these conditions, suggesting that contraction would be potentiated by ablation of AMPK. Indeed, in aortic rings from mice in which alpha1, the major catalytic subunit isoform in arterial smooth muscle, had been deleted, KCl- or phenylephrine-induced contraction was increased. The findings suggest that AMPK attenuates contraction by phosphorylating and inactivating MLCK. This might contribute to reduced ATP turnover in the tonic phase of smooth muscle contraction.
- Published
- 2008
- Full Text
- View/download PDF
114. A highly sensitive technique to measure myosin regulatory light chain phosphorylation: the first quantification in renal arterioles.
- Author
-
Takeya K, Loutzenhiser K, Shiraishi M, Loutzenhiser R, and Walsh MP
- Subjects
- Animals, Arterioles metabolism, Kidney metabolism, Lasers, Luminescence, Phosphorylation, Rats, Rats, Wistar, Electrophoresis, Polyacrylamide Gel methods, Isoelectric Focusing methods, Kidney blood supply, Muscle, Smooth, Vascular metabolism, Myosin Light Chains metabolism
- Abstract
Phosphorylation of the 20-kDa myosin regulatory light chains (LC(20)) plays a key role in the regulation of smooth muscle contraction. The level of LC(20) phosphorylation is governed by the relative activities of myosin light chain kinase and phosphatase pathways. The regulation of these two pathways differs in different smooth muscle types and in the actions of different vasoactive stimuli. Little is known concerning the regulation of LC(20) phosphorylation in the renal microcirculation. The available pharmacological probes are often nonspecific, and current techniques to directly measure LC(20) phosphorylation are not sensitive enough for quantification in small arterioles. We describe here a novel approach to address this important issue. Using SDS-PAGE with polyacrylamide-bound Mn(2+)-phosphate-binding tag and enhanced Western blot analysis, we were able to detect LC(20) phosphorylation using as little as 5 pg (250 amol) of isolated LC(20). Phosphorylated and unphosphorylated LC(20) were detected in single isolated afferent arterioles, and LC(20) phosphorylation levels could be accurately quantified in pooled samples of three arterioles (<300 cells). The phosphorylation level of LC(20) in the afferent arteriole was 6.8 +/- 1.7% under basal conditions and increased to 34.7 +/- 5.1% and 44.6 +/- 6.6% in response to 30 mM KCl and 10(-8) M angiotensin II, respectively. The application of this technique will enable investigations of the different determinants of LC(20) phosphorylation in afferent and efferent arterioles and provide insights into the signaling pathways that regulate LC(20) phosphorylation in the renal microvasculature under physiological and pathophysiological conditions.
- Published
- 2008
- Full Text
- View/download PDF
115. Up4A stimulates endothelium-independent contraction of isolated rat pulmonary artery.
- Author
-
Gui Y, Walsh MP, Jankowski V, Jankowski J, and Zheng XL
- Subjects
- Adenosine Triphosphate pharmacology, Animals, Endothelium, Vascular drug effects, In Vitro Techniques, Male, Pulmonary Artery drug effects, RNA genetics, RNA isolation & purification, Rats, Rats, Sprague-Dawley, Receptors, Purinergic P2 genetics, Receptors, Purinergic P2Y2, Reverse Transcriptase Polymerase Chain Reaction, Uridine Diphosphate pharmacology, Uridine Triphosphate pharmacology, Vasodilation drug effects, Vasodilator Agents pharmacology, Dinucleoside Phosphates pharmacology, Endothelium, Vascular physiology, Pulmonary Artery physiology
- Abstract
Extracellular nucleotides, such as ATP, UDP, and UTP, regulate pulmonary vascular tone through P2X and P2Y receptors. Recently, uridine adenosine tetraphosphate (Up(4)A) was reported as a novel endothelium-derived vasoconstrictive factor. Up(4)A contains both purine and pyrimidine moieties, which potentially activate P2X and P2Y receptors. The present study examined the effect of Up(4)A on contractility of isolated rat pulmonary artery. Up(4)A at 1-100 microM stimulated contraction in a concentration-dependent manner. Up(4)A was equipotent as UTP and UDP in the endothelium-denuded artery while much more effective than UTP and UDP in endothelium-intact preparations. The vasoconstrictor effect of Up(4)A was inhibited by suramin but not IP(5)I or desensitization of P2X receptors with alpha,beta-methylene-ATP (alpha,beta-Me-ATP). Up(4)A-induced contraction was also inhibited by pretreatment with thapsigargin, nitrendipine, or EGTA but unaffected by H1152. Furthermore, unlike ATP and UTP, Up(4)A did not induce relaxation of endothelium-intact preparations precontracted with phenylephrine. These results suggest that Up(4)A is a potent vasoconstrictor, but not a vasodilator, of the rat pulmonary artery. Up(4)A likely acts through a suramin-sensitive P2Y receptor. The contractile effect of Up(4)A involves the entry of extracellular Ca(2+) and release of Ca(2+) from intracellular stores but not Ca(2+) sensitization via the RhoA/Rho kinase pathway. Up(4)A, therefore, potentially plays an important role in the regulation of pulmonary vascular tone.
- Published
- 2008
- Full Text
- View/download PDF
116. Ancillary benefits for climate change mitigation and air pollution control in the world's motor vehicle fleets.
- Author
-
Walsh MP
- Subjects
- Air Pollution adverse effects, Air Pollution legislation & jurisprudence, Gasoline standards, Global Health, Humans, International Cooperation, Motor Vehicles statistics & numerical data, Vehicle Emissions legislation & jurisprudence, Air Pollution prevention & control, Greenhouse Effect, Motor Vehicles standards, Vehicle Emissions prevention & control
- Abstract
The global motor vehicle population has grown very rapidly in the past half century and is expected to continue to grow rapidly for the next several decades, especially in developing countries. As a result, vehicles are a major source of urban air pollution in many cities and are the fastest-growing source of greenhouse emissions. Strategies exist to reduce both problems, but many countries emphasize one over the other rather than pursuing strategies that reduce both concerns. Using diesel as an example, this article illustrates that it is now possible not only to reduce carbon dioxide with the increased use of diesel vehicles but also to improve urban air pollution. Doing so requires both stringent emissions regulations and clean fuels. Several principles contained in the Bellagio Memorandum are highlighted as guides for policy makers.
- Published
- 2008
- Full Text
- View/download PDF
117. The global experience with lead in gasoline and the lessons we should apply to the use of MMT.
- Author
-
Walsh MP
- Subjects
- Air Pollutants analysis, Environmental Exposure, Global Health, Humans, Fossil Fuels toxicity, Lead analysis, Organometallic Compounds adverse effects
- Abstract
As leaded gasoline is on the verge of being eliminated from all gasoline worldwide another neurotoxin has been put on the market as a lead substitute. Initial concerns regarding the potential adverse health impacts of this additive have emerged and some evidence indicates that its use could damage pollution controls on advance gasoline fueled vehicles. This paper attempts to draw upon the experience with lead in gasoline to see what lessons we should have learned and how these lessons should apply to the use of the new additive, methylcyclopentadienyl manganese tricarbonyl (MMT). Based on the global experience with lead, the question is whether we have sufficient information to conclude that the precautionary principle should apply to the use of MMT; this would require the developer of the additive to demonstrate conclusively that it is safe before it is sold around the world., ((c) 2007 Wiley-Liss, Inc.)
- Published
- 2007
- Full Text
- View/download PDF
118. Functional consequence of protein kinase A-dependent phosphorylation of the cardiac ryanodine receptor: sensitization of store overload-induced Ca2+ release.
- Author
-
Xiao B, Tian X, Xie W, Jones PP, Cai S, Wang X, Jiang D, Kong H, Zhang L, Chen K, Walsh MP, Cheng H, and Chen SR
- Subjects
- Animals, Cell Line, Heart Ventricles pathology, Humans, Isoproterenol pharmacology, Models, Biological, Mutation, Myocytes, Cardiac metabolism, Phosphorylation, Rats, Serine chemistry, Calcium metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Myocardium metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
The phosphorylation of the cardiac Ca(2+)-release channel (ryanodine receptor, RyR2) by protein kinase A (PKA) has been extensively characterized, but its functional consequence remains poorly defined and controversial. We have previously shown that RyR2 is phosphorylated by PKA at two major sites, serine 2,030 and serine 2,808, of which Ser-2,030 is the major PKA site responding to beta-adrenergic stimulation. Here we investigated the effect of the phosphorylation of RyR2 by PKA on the properties of single channels and on spontaneous Ca(2+) release during sarcoplasmic reticulum Ca(2+) overload, a process we have referred to as store overload-induced Ca(2+) release (SOICR). We found that PKA activated single RyR2 channels in the presence, but not in the absence, of luminal Ca(2+). On the other hand, PKA had no marked effect on the sensitivity of the RyR2 channel to activation by cytosolic Ca(2+). Importantly, the S2030A mutation, but not mutations of Ser-2,808, diminished the effect of PKA on RyR2. Furthermore, a phosphomimetic mutation, S2030D, potentiated the response of RyR2 to luminal Ca(2+) and enhanced the propensity for SOICR in HEK293 cells. In intact rat ventricular myocytes, the activation of PKA by isoproterenol reduced the amplitude and increased the frequency of SOICR. Confocal line-scanning fluorescence microscopy further revealed that the activation of PKA by isoproterenol increased the rate of Ca(2+) release and the propagation velocity of spontaneous Ca(2+) waves, despite reduced wave amplitude and resting cytosolic Ca(2+). Collectively, our data indicate that PKA-dependent phosphorylation enhances the response of RyR2 to luminal Ca(2+) and reduces the threshold for SOICR and that this effect of PKA is largely mediated by phosphorylation at Ser-2,030.
- Published
- 2007
- Full Text
- View/download PDF
119. Characterization of protein kinase pathways responsible for Ca2+ sensitization in rat ileal longitudinal smooth muscle.
- Author
-
Ihara E, Moffat L, Ostrander J, Walsh MP, and MacDonald JA
- Subjects
- Amides pharmacology, Animals, Apoptosis Regulatory Proteins metabolism, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Death-Associated Protein Kinases, Ileum enzymology, Indoles pharmacology, Maleimides pharmacology, Microcystins pharmacology, Mitogen-Activated Protein Kinases metabolism, Muscle Contraction drug effects, Muscle Proteins physiology, Muscle, Smooth enzymology, Phosphoproteins physiology, Protein Kinase C physiology, Protein Phosphatase 1 metabolism, Pyridines pharmacology, Rats, Calcium Signaling physiology, Ileum physiology, Muscle, Smooth physiology, Myosin Light Chains metabolism, Myosin-Light-Chain Phosphatase metabolism
- Abstract
We investigated the protein kinases responsible for myosin regulatory light chain (LC20) phosphorylation and regulation of myosin light chain phosphatase (MLCP) activity during microcystin (phosphatase inhibitor)-induced contraction at low Ca2+ concentrations of rat ileal smooth muscle stretched in the longitudinal axis. Application of 1 microM microcystin induced LC20 diphosphorylation and contraction of beta-escin-permeabilized rat ileal smooth muscle at pCa 9. The PKC inhibitor GF-109203x, the MEK inhibitor PD-98059, and the p38 MAPK inhibitor SB-203580 significantly reduced this contraction. These inhibitory effects were abolished when the microcystin concentration was increased to 10 muM, indicating that application of these kinase inhibitors generated an increase in MLCP activity. GF-109203x and PD-98059, but not SB-203580, significantly decreased the phosphorylation level of the myosin-targeting subunit of MLCP, MYPT1, at Thr-697 (rat sequence) during microcystin-induced contraction at pCa 9. On the other hand, SB-203580, but not GF-109203x or PD-98059, significantly reduced the phosphorylation level of the PKC-potentiated phosphatase inhibitor of 17 kDa (CPI-17). A zipper-interacting protein kinase (ZIPK) inhibitor (SM1 peptide) and a Rho-associated kinase inhibitor (Y-27632) had little effect on microcystin-induced contraction at pCa 9. In conclusion, PKC, ERK1/2, and p38 MAPK pathways facilitate microcystin-induced contraction at low Ca2+ concentrations by contributing to the inhibition of MLCP activity either through phosphorylation of MYPT1 or CPI-17 [probably mediated by integrin-linked kinase (ILK)]. ILK and not ZIPK is likely to be the protein kinase responsible for LC20 diphosphorylation during microcystin-induced contraction of rat ileal smooth muscle at pCa 9, similar to its recently described role in vascular smooth muscle. The negative regulation of MLCP by PKC and MAPKs during microcystin-induced contraction at pCa 9, which is not observed in vascular smooth muscle, may be unique to phasic smooth muscle.
- Published
- 2007
- Full Text
- View/download PDF
120. Predisposition to tetraploidy in pulmonary vascular smooth muscle cells derived from the Eker rats.
- Author
-
Gui Y, He GH, Walsh MP, and Zheng XL
- Subjects
- Animals, Cell Cycle, Centrosome metabolism, Cyclin B genetics, Cyclin E genetics, Cyclin-Dependent Kinases genetics, DNA biosynthesis, Female, Male, Mitogen-Activated Protein Kinases metabolism, Muscle, Smooth, Vascular enzymology, Myocytes, Smooth Muscle cytology, Myocytes, Smooth Muscle enzymology, Protein Kinases metabolism, Rats, Rats, Inbred Strains, Rats, Long-Evans, Ribosomal Protein S6 Kinases metabolism, TOR Serine-Threonine Kinases, Tuberous Sclerosis Complex 2 Protein, Tumor Suppressor Proteins metabolism, Up-Regulation genetics, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Polyploidy, Pulmonary Artery cytology, Pulmonary Artery metabolism
- Abstract
Somatic mutations in the tuberous sclerosis complex-2 (TSC2) gene are associated with pulmonary lymphangioleiomyomatosis (LAM), a disorder characterized by benign lesions of smooth muscle and/or smooth muscle-like cells in the lung. However, the cellular mechanisms underlying LAM disease are largely unknown. Given that the TSC2 gene product tuberin is involved in the regulation of cell growth and proliferation, the present study was designed to investigate the potential roles of TSC2 in regulation of the cell cycle. We studied cell cycle profiles of pulmonary vascular smooth muscle cells (SMCs) derived from Eker rats (Tsc2(+/EK)), a genetic model carrying a germline insertional deletion in one copy of the Tsc2 gene, and the wild-type rats (Tsc2(+/+)), a noncarrier counterpart. We found that Tsc2(+/EK), but not Tsc2(+/+), SMCs displayed increases in cells with > or =4N DNA content (> or =4N cells) and in the bromodeoxyuridine (BrdU) incorporation of > or =4N cells. Centrosome number was also increased in Tsc2(+/EK) SMCs, but the mitotic index was comparable between Tsc2(+/+) and Tsc2(+/EK) SMCs. Furthermore, Tsc2(+/EK) SMCs showed elevated phosphorylation of p70S6K and increased expression of cell cycle regulatory proteins Cdk1, cyclin B, Cdk2, and cyclin E. Inhibition of the mammalian target of rapamycin (mTOR) pathway by rapamycin not only inhibited the phosphorylation of p70(S6K) and the expression of cell cycle regulatory proteins but also reduced accumulation of > or =4N cells and BrdU incorporation of >4N cells. Therefore, our results demonstrate that Tsc2(+/EK) SMCs are predisposed to undergo tetraploidization, involving activation of the mTOR pathway. These findings suggest an important role of Tsc2 in regulation of the cell cycle.
- Published
- 2007
- Full Text
- View/download PDF
121. Structures and metal-ion-binding properties of the Ca2+-binding helix-loop-helix EF-hand motifs.
- Author
-
Gifford JL, Walsh MP, and Vogel HJ
- Subjects
- Amino Acid Sequence, Calbindins, Calmodulin chemistry, Calmodulin metabolism, Magnesium metabolism, Models, Biological, Models, Molecular, Protein Interaction Mapping, S100 Calcium Binding Protein G chemistry, S100 Calcium Binding Protein G metabolism, Thermodynamics, Calcium metabolism, EF Hand Motifs physiology
- Abstract
The 'EF-hand' Ca2+-binding motif plays an essential role in eukaryotic cellular signalling, and the proteins containing this motif constitute a large and functionally diverse family. The EF-hand is defined by its helix-loop-helix secondary structure as well as the ligands presented by the loop to bind the Ca2+ ion. The identity of these ligands is semi-conserved in the most common (the 'canonical') EF-hand; however, several non-canonical EF-hands exist that bind Ca2+ by a different co-ordination mechanism. EF-hands tend to occur in pairs, which form a discrete domain so that most family members have two, four or six EF-hands. This pairing also enables communication, and many EF-hands display positive co-operativity, thereby minimizing the Ca2+ signal required to reach protein saturation. The conformational effects of Ca2+ binding are varied, function-dependent and, in some cases, minimal, but can lead to the creation of a protein target interaction site or structure formation from a molten-globule apo state. EF-hand proteins exhibit various sensitivities to Ca2+, reflecting the intrinsic binding ability of the EF-hand as well as the degree of co-operativity in Ca2+ binding to paired EF-hands. Two additional factors can influence the ability of an EF-hand to bind Ca2+: selectivity over Mg2+ (a cation with very similar chemical properties to Ca2+ and with a cytoplasmic concentration several orders of magnitude higher) and interaction with a protein target. A structural approach is used in this review to examine the diversity of family members, and a biophysical perspective provides insight into the ability of the EF-hand motif to bind Ca2+ with a wide range of affinities.
- Published
- 2007
- Full Text
- View/download PDF
122. The functional consequence of RhoA knockdown by RNA interference in rat cerebral arteries.
- Author
-
Corteling RL, Brett SE, Yin H, Zheng XL, Walsh MP, and Welsh DG
- Subjects
- Animals, Female, In Vitro Techniques, Rats, Rats, Sprague-Dawley, rhoA GTP-Binding Protein genetics, Actins metabolism, Cerebral Arteries physiology, Gene Silencing, RNA Interference, Vasoconstriction physiology, rhoA GTP-Binding Protein metabolism
- Abstract
Uridine triphosphate (UTP) constricts cerebral arteries by activating transduction pathways that increase cytosolic [Ca(2+)] and myofilament Ca(2+) sensitivity. The signaling proteins that comprise these pathways remain uncertain with recent studies implicating a role for several G proteins. To start clarifying which G proteins enable UTP-induced vasoconstriction, a small interfering RNA (siRNA) approach was developed to knock down specified targets in rat cerebral arteries. siRNA directed against G(q) and RhoA was introduced into isolated cerebral arteries using reverse permeabilization. Following a defined period of organ culture, arteries were assayed for contractile function, mRNA levels, and protein expression. Targeted siRNA reduced RhoA or G(q) mRNA expression by 60-70%, which correlated with a reduction in RhoA but not G(q) protein expression. UTP-induced constriction was abolished in RhoA-depleted arteries, but this was not due to a reduction in myosin light chain phosphorylation. UTP-induced actin polymerization was attenuated in RhoA-depleted arteries, which would explain the loss of agonist-induced constriction. In summary, this study illustrates that siRNA approaches can be effectively used on intact arteries to induce targeted knockdown given that the protein turnover rate is sufficiently high. It also demonstrates that the principal role of RhoA in agonist-induced constriction is to facilitate the formation of F-actin, the physical structure to which phosphorylated myosin binds to elicit arterial constriction.
- Published
- 2007
- Full Text
- View/download PDF
123. Inhibition of zipper-interacting protein kinase function in smooth muscle by a myosin light chain kinase pseudosubstrate peptide.
- Author
-
Ihara E, Edwards E, Borman MA, Wilson DP, Walsh MP, and MacDonald JA
- Subjects
- Amino Acid Sequence, Animals, Binding, Competitive, Calcium metabolism, Calmodulin metabolism, Cell Line, Dose-Response Relationship, Drug, Humans, Ileum enzymology, Ileum metabolism, In Vitro Techniques, MAP Kinase Kinase Kinases, Models, Biological, Molecular Sequence Data, Muscle Contraction drug effects, Myocytes, Smooth Muscle enzymology, Myocytes, Smooth Muscle metabolism, Myosin-Light-Chain Kinase metabolism, Peptides metabolism, Phosphorylation, Protein Kinase Inhibitors metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Structure, Tertiary, Rats, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins metabolism, Transfection, Ileum drug effects, Myocytes, Smooth Muscle drug effects, Myosin Light Chains metabolism, Myosin-Light-Chain Kinase antagonists & inhibitors, Peptides pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
As a regulator of smooth muscle contractility, zipper-interacting protein kinase (ZIPK) appears to phosphorylate the regulatory myosin light chain (RLC20), directly or indirectly, at Ser19 and Thr18 in a Ca(2+)-independent manner. The calmodulin-binding and autoinhibitory domain of myosin light chain kinase (MLCK) shares similarity to a sequence found in ZIPK. This similarity in sequence prompted an investigation of the SM1 peptide, which is derived from the autoinhibitory region of MLCK, as a potential inhibitor of ZIPK. In vitro studies showed that SM1 is a competitive inhibitor of a constitutively active 32-kDa form of ZIPK with an apparent K(i) value of 3.4 microM. Experiments confirmed that the SM1 peptide is also active against full-length ZIPK. In addition, ZIPK autophosphorylation was reduced by SM1. ZIPK activity is independent of calmodulin; however, calmodulin suppressed the in vitro inhibitory potential of SM1, likely as a result of nonspecific binding of the peptide to calmodulin. Treatment of ileal smooth muscle with exogenous ZIPK was accompanied by an increase in RLC20 diphosphorylation, distinguishing between ZIPK [and integrin-linked kinase (ILK)] and MLCK actions. Administration of SM1 suppressed steady-state muscle tension developed by the addition of exogenous ZIPK to Triton-skinned rat ileal muscle strips with or without calmodulin depletion by trifluoperazine. The decrease in contractile force was associated with decreases in both RLC20 mono- and diphosphorylation. In summary, we present the SM1 peptide as a novel inhibitor of ZIPK. We also conclude that the SM1 peptide, which has no effect on ILK, can be used to distinguish between ZIPK and ILK effects in smooth muscle tissues.
- Published
- 2007
- Full Text
- View/download PDF
124. Identification of a novel interaction between the Ca(2+)-binding protein S100A11 and the Ca(2+)- and phospholipid-binding protein annexin A6.
- Author
-
Chang N, Sutherland C, Hesse E, Winkfein R, Wiehler WB, Pho M, Veillette C, Li S, Wilson DP, Kiss E, and Walsh MP
- Subjects
- Amino Acid Sequence, Animals, Annexin A6 chemistry, Annexin A6 genetics, Chickens, Immunohistochemistry, In Vitro Techniques, Liposomes, Male, Molecular Sequence Data, Muscle, Smooth, Vascular metabolism, Phospholipids chemistry, Protein Binding, Protein Structure, Tertiary, Rats, Rats, Sprague-Dawley, Recombinant Proteins chemistry, S100 Proteins chemistry, S100 Proteins genetics, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Annexin A6 metabolism, Calcium metabolism, Muscle, Smooth metabolism, Phospholipids metabolism, S100 Proteins metabolism
- Abstract
S100A11 is a member of the S100 family of EF-hand Ca(2+)-binding proteins, which is expressed in smooth muscle and other tissues. Ca(2+) binding to S100A11 induces a conformational change that exposes a hydrophobic surface for interaction with target proteins. Affinity chromatography with immobilized S100A11 was used to isolate a 70-kDa protein from smooth muscle that bound to S100A11 in a Ca(2+)-dependent manner and was identified by mass spectrometry as annexin A6. Direct Ca(2+)-dependent interaction between S100A11 and annexin A6 was confirmed by affinity chromatography of the purified bacterially expressed proteins, by gel overlay of annexin A6 with purified S100A11, by chemical cross-linking, and by coprecipitation of S100A11 with annexin A6 bound to liposomes. The expression of S100A11 and annexin A6 in the same cell type was verified by RT-PCR and immunocytochemistry of isolated vascular smooth muscle cells. The site of binding of S100A11 on annexin A6 was investigated by partial tryptic digestion and deletion mutagenesis. The unique NH(2) terminal head region of annexin A6 was not required for S100A11 binding, but binding sites were identified in both NH(2)- and COOH-terminal halves of the molecule. We hypothesize that an agonist-induced increase in cytosolic free [Ca(2+)] leads to formation of a complex of S100A11 and annexin A6, which forms a physical connection between the plasma membrane and the cytoskeleton, or plays a role in the formation of signaling complexes at the level of the sarcolemma.
- Published
- 2007
- Full Text
- View/download PDF
125. Endoreduplication of human smooth muscle cells induced by 2-methoxyestradiol: a role for cyclin-dependent kinase 2.
- Author
-
Gui Y, Yin H, He JY, Yang SH, Walsh MP, and Zheng XL
- Subjects
- 2-Methoxyestradiol, Aneuploidy, Aorta, Cell Division drug effects, DNA Replication drug effects, Estradiol pharmacology, Gene Expression Regulation, Enzymologic, Humans, Hypertension pathology, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular enzymology, Cyclin-Dependent Kinase 2 genetics, Estradiol analogs & derivatives, Muscle, Smooth, Vascular cytology
- Abstract
Endoreduplication has been suggested to contribute to the development of hypertrophy of smooth muscle cells (SMCs) in hypertension. However, endoreduplication in vascular SMCs and the underlying molecular mechanisms are not clear. Treatment of human SMCs with 10 microM 2-methoxyestradiol (2-ME) for 24 h induces accumulation of cells with > or =4N DNA content, and some polyploid/aneuploid cells actively synthesize their DNA, suggesting the occurrence of endoreduplication. In addition, 2-ME treatment upregulates the expression of cyclin-dependent kinase 2 (Cdk2). The present study was designed to characterize endoreduplication of human SMCs and explore the potential roles of Cdk2 in endoreduplication induced by 2-ME. Treatment with 2-ME (10 microM) for 2-4 days not only caused increases in >4N cells and their reentry into S phase but also induced overduplication of chromosomes. Furthermore, 2-ME increased the kinase activity of Cdk2 and its interaction with cyclin E. Inducible overexpression of dominant-negative Cdk2 in human SMCs inhibited both DNA synthesis of >4N cells and the accumulation of >4N cells induced by 2-ME. We conclude that 2-ME induces endoreduplication of human SMCs and Cdk2 plays an important role in endoreduplication in response to 2-ME.
- Published
- 2007
- Full Text
- View/download PDF
126. Phosphorylation of the protein phosphatase type 1 inhibitor protein CPI-17 by protein kinase C.
- Author
-
Walsh MP, Susnjar M, Deng J, Sutherland C, Kiss E, and Wilson DP
- Subjects
- Animals, Calcium metabolism, Myosin-Light-Chain Kinase metabolism, Myosin-Light-Chain Phosphatase metabolism, Phosphorylation, Swine, Phosphoprotein Phosphatases metabolism, Phosphoproteins metabolism, Protein Kinase C metabolism
- Abstract
CPI-17 is a cytosolic protein of 17 kDa that becomes a potent inhibitor of certain type 1 protein serine/threonine phosphatases, including smooth muscle myosin light-chain phosphatase (MLCP), when phosphorylated at Thr38. Several protein kinases are capable of phosphorylating CPI-17 at this site in vitro; however, in intact tissue, compelling evidence only exists for phosphorylation by protein kinase C (PKC). Agonist-induced activation of heterotrimeric G proteins of the Gq/11 family via seven-transmembrane domain-containing, G protein-coupled receptors results in phospholipase Cbeta-mediated hydrolysis of membrane phosphatidylinositol 4,5-bisphosphate to generate inositol 1,4,5-trisphosphate (IP3) and 1,2-diacylglycerol (DAG). IP3 triggers Ca2+ release from the sarcoplasmic reticulum. DAG and Ca2+ together activate classical isoforms of PKC, and DAG activates novel PKC isoforms without a requirement for Ca2+. Activated PKC phosphorylates CPI-17 at Thr38, enhancing its potency of inhibition of MLCP approx 1000-fold. The myosin light-chain kinase (MLCK):MLCP activity ratio is thereby increased at the prevailing cytosolic free-Ca2+ concentration ([Ca2+]i), resulting in an increase in phosphorylation of the 20-kDa light chains of myosin II (LC20) catalyzed by Ca2+- and calmodulin-dependent MLCK and contraction of the smooth muscle. Physiologically, this mechanism can account for some instances of Ca2+ sensitization of smooth muscle contraction (i.e., an increase in force in response to agonist stimulation without a change in [Ca2+]i).
- Published
- 2007
- Full Text
- View/download PDF
127. Localization of telokin at the intercalated discs of cardiac myocytes.
- Author
-
Kargacin GJ, Hunt D, Emmett T, Rokolya A, McMartin GA, Wirch E, Walsh MP, Ikebe M, and Kargacin ME
- Subjects
- Animals, Cells, Cultured, Myocytes, Cardiac ultrastructure, Peptide Fragments, Rats, Myocytes, Cardiac metabolism, Myosin-Light-Chain Kinase metabolism, Peptides metabolism, Sarcomeres metabolism, Subcellular Fractions metabolism
- Abstract
Telokin is identical in sequence to the C-terminal portion of myosin light chain kinase but is expressed independently. We have used monoclonal antibodies specific to the non-telokin portion of myosin light chain kinase and to telokin, immunofluorescence microscopy and image reconstruction to demonstrate the presence of telokin in cardiac myocytes and to study its subcellular distribution. Antibodies to telokin labeled the intercalated discs of adult cardiac myocytes and similar structures in isolated intercalated disc preparations. Antibodies specific to the non-telokin portion of myosin light chain kinase did not label intercalated discs in either of these preparations. Western blots of isolated intercalated discs with anti-telokin revealed a 23kDa protein that co-migrates with purified telokin on SDS-PAGE. Deconvolution, reconstruction and analysis of fluorescence images of isolated intercalated discs labeled with anti-telokin and anti-beta-catenin, anti-gamma-catenin or anti-connexin43 indicated that telokin is only partially co-localized with these proteins at the discs.
- Published
- 2006
- Full Text
- View/download PDF
128. The role of adenosine in the early respiratory and cardiovascular changes evoked by chronic hypoxia in the rat.
- Author
-
Walsh MP and Marshall JM
- Subjects
- Animals, Blood Pressure, Enzyme Inhibitors pharmacology, Guanidines pharmacology, Heart Rate, Hemodynamics, Hypoxia, Male, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide Synthase antagonists & inhibitors, Rats, Rats, Wistar, Respiratory Function Tests, Theophylline analogs & derivatives, Theophylline pharmacology, Xanthines pharmacology, Adenosine pharmacology, Cardiovascular System drug effects, Receptor, Adenosine A1 drug effects, Respiration drug effects
- Abstract
Experiments were performed on anaesthetized normoxic (N) rats and chronically hypoxic rats that had been exposed to 12% O2 for 1, 3 or 7 days (1, 3 or 7CH rats). The adenosine A1 receptor antagonist DPCPX did not affect the resting hyperventilation of 1-7CH rats breathing 12% O2 and increased resting heart rate (HR) in 1CH rats only. DPCPX partially restored the decreased baseline arterial pressure (ABP) and increased femoral vascular conductance (FVC) of 1 and 3CH rats, but had no effect in N or 7CH rats. DPCPX also attenuated the decrease in arterial blood pressure (ABP) and increase in FVC evoked by acute hypoxia in N and 1-7CH rats. The non-selective adenosine receptor antagonist 8-SPT had no further effect on baselines or cardiovascular responses to acute hypoxia, but attenuated the hypoxia-evoked increase in respiratory frequency in 1-7CH rats. In N, and 1 and 3CH rats, the inducible nitric oxide synthase (iNOS) inhibitor aminoguanidine had no effect on baselines or increases in FVC evoked by acetylcholine. We propose: (i) that tonically released adenosine acting on A1 receptors reduces HR in 1CH rats and stimulates endothelial NOS in 1 and 3CH rats to decrease ABP and increase FVC, the remaining NO-dependent tonic vasodilatation being independent of iNOS activity; (ii) that in 7CH rats, tonic adenosine release has waned; (iii) that in 1-7CH rats, adenosine released by acute hypoxia stimulates A1 but not A2 receptors to produce muscle vasodilatation, and stimulates carotid body A2 receptors to increase respiration.
- Published
- 2006
- Full Text
- View/download PDF
129. The early effects of chronic hypoxia on the cardiovascular system in the rat: role of nitric oxide.
- Author
-
Walsh MP and Marshall JM
- Subjects
- Animals, Blood Pressure, Calcitonin Gene-Related Peptide pharmacology, Cardiovascular System drug effects, Cardiovascular System physiopathology, Enzyme Inhibitors pharmacology, Femoral Artery physiopathology, Heart Rate, Hypoxia physiopathology, Male, Muscle, Skeletal drug effects, Muscle, Skeletal enzymology, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide Synthase antagonists & inhibitors, Nitric Oxide Synthase metabolism, Rats, Rats, Wistar, Regional Blood Flow, Respiratory Function Tests, Time Factors, Cardiovascular System metabolism, Hypoxia metabolism, Muscle, Skeletal blood supply, Nitric Oxide metabolism, Vasodilation
- Abstract
Experiments were performed under Saffan anaesthesia on normoxic (N) rats and on chronically hypoxic rats exposed to 12% O2 for 1, 3 or 7 days (1, 3 or 7CH rats): N rats routinely breathed 21% O2 and CH rats 12% O2. The 1, 3 and 7CH rats showed resting hyperventilation relative to N rats, but baseline heart rate (HR) was unchanged and arterial blood pressure (ABP) was lowered. Femoral vascular conductance (FVC) was increased in 1 and 3CH rats, but not 7CH rats. When 1-7CH rats were acutely switched to breathing 21% O2 for 5 min, ABP increased and FVC decreased, consistent with removal of a hypoxic dilator stimulus that is waning in 7CH rats. We propose that this is because the increase in haematocrit and vascular remodelling in skeletal muscle help restore the O2 supply. The increases in FVC evoked by acute hypoxia (8% O2 for 5 min) and by infusion for 5 min of alpha-calcitonin gene-related peptide (alpha-CGRP), which are NO-dependent, were particularly accentuated in 1CH, relative to N rats. The NO synthesis inhibitor L-NAME increased ABP, decreased HR and greatly reduced FVC, and attenuated increases in FVC evoked by acute hypoxia and alpha-CGRP, such that baselines and responses were similar in N and 1-7CH rats. We propose that in the first few days of chronic hypoxia there is tonic NO-dependent vasodilatation in skeletal muscle that is associated with accentuated dilator responsiveness to acute hypoxia and dilator substances that are NO -dependent.
- Published
- 2006
- Full Text
- View/download PDF
130. Key role of Kv1 channels in vasoregulation.
- Author
-
Chen TT, Luykenaar KD, Walsh EJ, Walsh MP, and Cole WC
- Subjects
- Animals, Arteries physiology, Feedback, Physiological, Genes, Dominant, In Vitro Techniques, Kv1.5 Potassium Channel genetics, Male, Pressure, Rabbits, Rats, Rats, Sprague-Dawley, Transfection, Kv1.5 Potassium Channel physiology, Vasoconstriction physiology
- Abstract
Small arteries play an essential role in the regulation of blood pressure and organ-specific blood flow by contracting in response to increased intraluminal pressure, ie, the myogenic response. The molecular basis of the myogenic response remains to be defined. To achieve incremental changes in arterial diameter, as well as blood pressure or organ-specific blood flow, the depolarizing influence of intravascular pressure on vascular smooth muscle membrane potential that elicits myogenic contraction must be precisely controlled by an opposing hyperpolarizing influence. Here we use a dominant-negative molecular strategy and pressure myography to determine the role of voltage-dependent Kv1 potassium channels in vasoregulation, specifically, whether they act as a negative-feedback control mechanism of the myogenic response. Functional Kv1 channel expression was altered by transfection of endothelium-denuded rat middle cerebral arteries with cDNAs encoding c-myc epitope-tagged, dominant-negative mutant or wild-type rabbit Kv1.5 subunits. Expression of mutant Kv1.5 dramatically enhanced, whereas wild-type subunit expression markedly suppressed, the myogenic response over a wide range of intraluminal pressures. These effects on arterial diameter were associated with enhanced and reduced myogenic depolarization by mutant and wild-type Kv1.5 subunit expression, respectively. Expression of myc-tagged mutant and wild-type Kv1.5 subunit message and protein in transfected but not control arteries was confirmed, and isolated myocytes of transfected but not control arteries exhibited anti-c-myc immunofluorescence. No changes in message encoding other known, non-Kv1 elements of the myogenic response were apparent. These findings provide the first molecular evidence that Kv1-containing delayed rectifier K+ (K(DR)) channels are of fundamental importance for control of arterial diameter and, thereby, peripheral vascular resistance, blood pressure, and organ-specific blood flow.
- Published
- 2006
- Full Text
- View/download PDF
131. The CH-domain of calponin does not determine the modes of calponin binding to F-actin.
- Author
-
Galkin VE, Orlova A, Fattoum A, Walsh MP, and Egelman EH
- Subjects
- Actins ultrastructure, Animals, Calcium-Binding Proteins genetics, Calcium-Binding Proteins ultrastructure, Crystallography, X-Ray, Image Processing, Computer-Assisted, Microfilament Proteins genetics, Microfilament Proteins ultrastructure, Models, Molecular, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Protein Binding, Calponins, Actins metabolism, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins metabolism, Microfilament Proteins chemistry, Microfilament Proteins metabolism, Protein Conformation
- Abstract
Many actin-binding proteins have been observed to have a modular architecture. One of the most abundant modules is the calponin-homology (CH) domain, found as tandem repeats in proteins that cross-link actin filaments (such as fimbrin, spectrin and alpha-actinin) or link the actin cytoskeleton to intermediate filaments (such as plectin). In proteins such as the eponymous calponin, IQGAP1, and Scp1, a single CH-domain exists, but there has been some controversy over whether this domain binds to actin filaments. A previous three-dimensional reconstruction of the calponin-F-actin complex has led to the conclusion that the visualized portion of calponin bound to actin belongs to its amino-terminal homology (CH) domain. We show, using a calponin fragment lacking the CH-domain, that this domain is not bound to F-actin, and cannot be positioning calponin on F-actin as hypothesized. Further, using classification methods, we show a multiplicity in cooperative modes of binding of calponin to F-actin, similar to what has been observed for other actin-binding proteins such as tropomyosin and cofilin. Our results suggest that the form and function of the structurally conserved CH-domain found in many other actin-binding proteins have diverged. This has broad implications for inferring function from the presence of structurally conserved domains.
- Published
- 2006
- Full Text
- View/download PDF
132. Ser-2030, but not Ser-2808, is the major phosphorylation site in cardiac ryanodine receptors responding to protein kinase A activation upon beta-adrenergic stimulation in normal and failing hearts.
- Author
-
Xiao B, Zhong G, Obayashi M, Yang D, Chen K, Walsh MP, Shimoni Y, Cheng H, Ter Keurs H, and Chen SR
- Subjects
- Adrenergic beta-Agonists therapeutic use, Animals, Arrhythmias, Cardiac physiopathology, Benzylamines pharmacology, Blotting, Western, Calcium Signaling drug effects, Calcium Signaling physiology, Carrier Proteins pharmacology, Cell Line, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Heart Failure drug therapy, Humans, Ion Channel Gating drug effects, Ion Transport drug effects, Isoproterenol therapeutic use, Kidney cytology, Marine Toxins, Mice, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Oxazoles pharmacology, Peptide Fragments pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases physiology, Phosphorylation, Phosphoserine chemistry, Protein Serine-Threonine Kinases physiology, Rabbits, Rats, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins physiology, Ryanodine Receptor Calcium Release Channel genetics, Ryanodine Receptor Calcium Release Channel physiology, Sodium-Calcium Exchanger metabolism, Staurosporine pharmacology, Structure-Activity Relationship, Sulfonamides pharmacology, Transfection, Adrenergic beta-Agonists pharmacology, Cyclic AMP-Dependent Protein Kinases physiology, Heart Failure metabolism, Isoproterenol pharmacology, Protein Processing, Post-Translational, Ryanodine Receptor Calcium Release Channel chemistry
- Abstract
We have recently shown that RyR2 (cardiac ryanodine receptor) is phosphorylated by PKA (protein kinase A/cAMP-dependent protein kinase) at two major sites, Ser-2030 and Ser-2808. In the present study, we examined the properties and physiological relevance of phosphorylation of these two sites. Using site- and phospho-specific antibodies, we demonstrated that Ser-2030 of both recombinant and native RyR2 from a number of species was phosphorylated by PKA, indicating that Ser-2030 is a highly conserved PKA site. Furthermore, we found that the phosphorylation of Ser-2030 responded to isoproterenol (isoprenaline) stimulation in rat cardiac myocytes in a concentration- and time-dependent manner, whereas Ser-2808 was already substantially phosphorylated before beta-adrenergic stimulation, and the extent of the increase in Ser-2808 phosphorylation after beta-adrenergic stimulation was much less than that for Ser-2030. Interestingly, the isoproterenol-induced phosphorylation of Ser-2030, but not of Ser-2808, was markedly inhibited by PKI, a specific inhibitor of PKA. The basal phosphorylation of Ser-2808 was also insensitive to PKA inhibition. Moreover, Ser-2808, but not Ser-2030, was stoichiometrically phosphorylated by PKG (protein kinase G). In addition, we found no significant phosphorylation of RyR2 at the Ser-2030 PKA site in failing rat hearts. Importantly, isoproterenol stimulation markedly increased the phosphorylation of Ser-2030, but not of Ser-2808, in failing rat hearts. Taken together, these observations indicate that Ser-2030, but not Ser-2808, is the major PKA phosphorylation site in RyR2 responding to PKA activation upon beta-adrenergic stimulation in both normal and failing hearts, and that RyR2 is not hyperphosphorylated by PKA in heart failure. Our results also suggest that phosphorylation of RyR2 at Ser-2030 may be an important event associated with altered Ca2+ handling and cardiac arrhythmia that is commonly observed in heart failure upon beta-adrenergic stimulation.
- Published
- 2006
- Full Text
- View/download PDF
133. The force is with alpha-actinin: dynamic regulation of the extracellular matrix-cytoskeletal connection in airway smooth muscle.
- Author
-
Walsh MP
- Subjects
- Animals, Calcium metabolism, Cytoskeleton ultrastructure, Extracellular Matrix ultrastructure, Feedback physiology, Humans, Lung, Muscle, Smooth cytology, Rats, Stress, Mechanical, Actinin metabolism, Cytoskeleton physiology, Extracellular Matrix physiology, Muscle Contraction physiology, Muscle, Smooth physiology, Pulmonary Alveoli cytology, Pulmonary Alveoli physiology
- Published
- 2006
- Full Text
- View/download PDF
134. Integrin-linked kinase is responsible for Ca2+-independent myosin diphosphorylation and contraction of vascular smooth muscle.
- Author
-
Wilson DP, Sutherland C, Borman MA, Deng JT, Macdonald JA, and Walsh MP
- Subjects
- Animals, Calcium pharmacology, MAP Kinase Kinase Kinases, Microcystins, Muscle Contraction drug effects, Muscle Contraction physiology, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Peptides, Cyclic pharmacology, Phosphorylation drug effects, Rats, Calcium metabolism, Muscle, Smooth, Vascular enzymology, Muscle, Smooth, Vascular physiology, Myosins metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Smooth muscle contraction is activated by phosphorylation at Ser-19 of LC20 (the 20 kDa light chains of myosin II) by Ca2+/calmodulin-dependent MLCK (myosin light-chain kinase). Diphosphorylation of LC20 at Ser-19 and Thr-18 is observed in smooth muscle tissues and cultured cells in response to various contractile stimuli, and in pathological circumstances associated with hypercontractility. MLCP (myosin light-chain phosphatase) inhibition can lead to LC20 diphosphorylation and Ca2+-independent contraction, which is not attributable to MLCK. Two kinases have emerged as candidates for Ca2+-independent LC20 diphosphorylation: ILK (integrin-linked kinase) and ZIPK (zipper-interacting protein kinase). Triton X-100-skinned rat caudal arterial smooth muscle was used to investigate the relative importance of ILK and ZIPK in Ca2+-independent, microcystin (phosphatase inhibitor)-induced LC20 diphosphorylation and contraction. Western blotting and in-gel kinase assays revealed that both kinases were retained in this preparation. Ca2+-independent contraction of calmodulin-depleted tissue in response to microcystin was resistant to MLCK inhibitors [AV25 (a 25-amino-acid peptide derived from the autoinhibitory domain of MLCK), ML-7, ML-9 and wortmannin], protein kinase C inhibitor (GF109203X) and Rho-associated kinase inhibitors (Y-27632 and H-1152), but blocked by the non-selective kinase inhibitor staurosporine. ZIPK was inhibited by AV25 (IC50 0.63+/-0.05 microM), whereas ILK was insensitive to AV25 (at concentrations as high as 100 microM). AV25 had no effect on Ca2+-independent, microcystin-induced LC20 mono- or di-phosphorylation, with a modest effect on force. We conclude that direct inhibition of MLCP in the absence of Ca2+ unmasks ILK activity, which phosphorylates LC20 at Ser-19 and Thr-18 to induce contraction. ILK is probably the kinase responsible for myosin diphosphorylation in vascular smooth muscle cells and tissues.
- Published
- 2005
- Full Text
- View/download PDF
135. Modulation of PGF2alpha- and hypoxia-induced contraction of rat intrapulmonary artery by p38 MAPK inhibition: a nitric oxide-dependent mechanism.
- Author
-
Knock GA, De Silva AS, Snetkov VA, Siow R, Thomas GD, Shiraishi M, Walsh MP, Ward JP, and Aaronson PI
- Subjects
- Animals, Calcium metabolism, Calcium Signaling drug effects, Constriction, Pathologic chemically induced, Constriction, Pathologic metabolism, Enzyme Inhibitors pharmacology, HSP27 Heat-Shock Proteins, Heat-Shock Proteins metabolism, Male, Muscle Contraction drug effects, Neoplasm Proteins metabolism, Nitric Oxide Synthase Type III antagonists & inhibitors, Nitric Oxide Synthase Type III metabolism, Organ Culture Techniques methods, Phosphorylation drug effects, Rats, Rats, Wistar, p38 Mitogen-Activated Protein Kinases antagonists & inhibitors, Dinoprost pharmacology, Hypoxia metabolism, Nitric Oxide metabolism, Oxytocics administration & dosage, Pulmonary Artery metabolism, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
The mechanisms through which p38 mitogen-activated protein kinase (p38 MAPK) is involved in smooth muscle contraction remain largely unresolved. We examined the role of p38 MAPK in prostaglandin F(2alpha) (PGF(2alpha))-induced vasoconstriction and in hypoxic pulmonary vasoconstriction (HPV) of rat small intrapulmonary arteries (IPA). The p38 MAPK inhibitors SB-203580 and SB-202190 strongly inhibited PGF(2alpha)-induced vasoconstriction, with IC(50)s of 1.6 and 1.2 microM, whereas the inactive analog SB-202474 was approximately 30-fold less potent. Both transient and sustained phases of HPV were suppressed by SB-203580, but not by SB-202474 (both 2 microM). Western blot analysis revealed that PGF(2alpha) (20 microM) increased phosphorylation of p38 MAPK and of heat shock protein 27 (HSP27), and this was abolished by SB-203580 but not by SB-202474 (both 2 microM). Endothelial denudation or blockade of endothelial nitric oxide (NO) synthase with N(omega)-nitro-L-arginine methyl ester (L-NAME) significantly suppressed the relaxation of PGF(2alpha)-constricted IPA by SB-203580, but not by SB-202474. Similarly, the inhibition of HPV by SB-203580 was prevented by prior treatment with L-NAME. SB-203580 (2 microM), but not SB-202474, enhanced relaxation-induced by the NO donor S-nitroso-N-acetylpenicillamine (SNAP) in endothelium-denuded IPA constricted with PGF(2alpha). In alpha-toxin-permeabilized IPA, SB-203580-induced relaxation occurred in the presence but not the absence of the NO donor sodium nitroprusside (SNP); SB-202474 was without effect even in the presence of SNP. In intact IPA, neither PGF(2alpha)- nor SNAP-mediated changes in cytosolic free Ca(2+) were affected by SB-203580. We conclude that p38 MAPK contributes to PGF(2alpha)- and hypoxia-induced constriction of rat IPA primarily by antagonizing the underlying Ca(2+)-desensitizing actions of NO.
- Published
- 2005
- Full Text
- View/download PDF
136. Augmentation of alpha1-adrenoceptor-mediated contraction by warming without increased phosphorylation of myosin in rat caudal arterial smooth muscle.
- Author
-
Mita M, Walsh MP, and Saito M
- Subjects
- Animals, Arteries drug effects, Arteries metabolism, Dose-Response Relationship, Drug, In Vitro Techniques, Male, Muscle Contraction drug effects, Muscle, Smooth, Vascular metabolism, Myosin Light Chains metabolism, Phosphorylation, Rats, Rats, Sprague-Dawley, Adrenergic alpha-Agonists pharmacology, Hot Temperature, Imidazoles pharmacology, Muscle, Smooth, Vascular drug effects
- Abstract
We previously reported the relationship between alpha1-adrenoceptor-mediated contraction and phosphorylation of 20-kDa myosin light chain (LC20) in de-endothelialized rat caudal arterial smooth muscle at room temperature (Mita M, Walsh MP. Biochem J. 1997;327:669-674). We now describe the effect of increasing the temperature to 37 degrees C on this relationship. The EC50 value (76.6 +/- 18.2 nM) for cirazoline (alpha1-adrenergic agonist)-induced contraction of the strips at room temperature (23 degrees C) was significantly greater than that (14.5 +/- 1.9 nM) at 37 degrees C. The initial rate of the contraction to a sub-maximal concentration of cirazoline (0.3 microM) was similar at the two temperatures. However, cirazoline-induced maximal force at 37 degrees C was approximately 1.8 times that at room temperature. LC20 phosphorylation in response to cirazoline at room temperature and 37 degrees C closely matched the time courses of contraction, but values were not significantly different at the two temperatures: resting phosphorylation levels were 0.09 +/- 0.04 mol P(i)/mol LC20 at 37 degrees C and 0.22 +/- 0.06 mol P(i)/mol LC20 at room temperature; maximal cirazoline-stimulated LC20 phosphorylation levels were 0.58 +/- 0.08 mol P(i)/mol LC20 at room temperature and 0.49 +/- 0.05 mol P(i)/mol LC20 at 37 degrees C. We conclude, therefore, that the enhanced cirazoline-induced contraction at 37 degrees C is not due to increased LC20 phosphorylation.
- Published
- 2005
- Full Text
- View/download PDF
137. Thromboxane A2-induced contraction of rat caudal arterial smooth muscle involves activation of Ca2+ entry and Ca2+ sensitization: Rho-associated kinase-mediated phosphorylation of MYPT1 at Thr-855, but not Thr-697.
- Author
-
Wilson DP, Susnjar M, Kiss E, Sutherland C, and Walsh MP
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Amino Acid Sequence, Animals, Arteries physiology, Calcium Channel Blockers pharmacology, Calcium Channels, L-Type, Carrier Proteins chemistry, Intracellular Signaling Peptides and Proteins, Muscle Proteins metabolism, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular physiology, Myosin-Light-Chain Kinase metabolism, Phosphoprotein Phosphatases chemistry, Phosphoproteins metabolism, Phosphorylation, Protein Phosphatase 1, Rats, Thromboxane A2 antagonists & inhibitors, Vasoconstriction drug effects, Vasoconstrictor Agents pharmacology, rho-Associated Kinases, Calcium physiology, Calcium Signaling, Carrier Proteins metabolism, Phosphoprotein Phosphatases metabolism, Protein Serine-Threonine Kinases metabolism, Thromboxane A2 physiology, Vasoconstriction physiology
- Abstract
The signal transduction pathway whereby the TxA2 (thromboxane A2) mimetic U-46619 activates vascular smooth muscle contraction was investigated in de-endothelialized rat caudal artery. U-46619-evoked contraction was inhibited by the TP receptor (TxA2 receptor) antagonist SQ-29548, the ROK (Rho-associated kinase) inhibitors Y-27632 and H-1152, the MLCK (myosin light-chain kinase) inhibitors ML-7, ML-9 and wortmannin, the voltagegated Ca2+-channel blocker nicardipine, and removal of extracellular Ca2+; the protein kinase C inhibitor GF109203x had no effect. U-46619 elicited Ca2+ sensitization in a-toxin-permeabilized tissue. U-46619 induced activation of the small GTPase RhoA, consistent with the involvement of ROK. Two downstream targets of ROK were investigated: CPI-17 [protein kinase C-potentiated inhibitory protein for PP1 (protein phosphatase type 1) of 17 kDa], a myosin light-chain phosphatase inhibitor, was not phosphorylated at the functional site (Thr-38); phosphorylation of MYPT1 (myosin-targeting subunit of myosin light-chain phosphatase) was significantly increased at Thr-855, but not Thr-697. U-46619-evoked contraction correlated with phosphorylation of the 20 kDa light chains of myosin. We conclude that: (i) U-46619 induces contraction via activation of the Ca2+/calmodulin/MLCK pathway and of the RhoA/ROK pathway; (ii) Thr-855 of MYPT1 is phosphorylated by ROK at rest and in response to U-46619 stimulation; (iii) Thr-697 of MYPT1 is phosphorylated by a kinase other than ROK under resting conditions, and is not increased in response to U-46619 treatment; and (iv) neither ROK nor protein kinase C phosphorylates CPI-17 in this vascular smooth muscle in response to U-46619.
- Published
- 2005
- Full Text
- View/download PDF
138. Characterization of a novel PKA phosphorylation site, serine-2030, reveals no PKA hyperphosphorylation of the cardiac ryanodine receptor in canine heart failure.
- Author
-
Xiao B, Jiang MT, Zhao M, Yang D, Sutherland C, Lai FA, Walsh MP, Warltier DC, Cheng H, and Chen SR
- Subjects
- Animals, Calcium metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Dogs, Isoproterenol pharmacology, Myocytes, Cardiac metabolism, Phosphorylation, Serine metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Heart Failure metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Hyperphosphorylation of the cardiac Ca2+ release channel (ryanodine receptor, RyR2) by protein kinase A (PKA) at serine-2808 has been proposed to be a key mechanism responsible for cardiac dysfunction in heart failure (HF). However, the sites of PKA phosphorylation in RyR2 and their phosphorylation status in HF are not well defined. Here we used various approaches to investigate the phosphorylation of RyR2 by PKA. Mutating serine-2808, which was thought to be the only PKA phosphorylation site in RyR2, did not abolish the phosphorylation of RyR2 by PKA. Two-dimensional phosphopeptide mapping revealed two major PKA phosphopeptides, one of which corresponded to the known serine-2808 site. Another, novel, PKA phosphorylation site, serine 2030, was identified by Edman sequencing. Using phospho-specific antibodies, we showed that the novel serine-2030 site was phosphorylated in rat cardiac myocytes stimulated with isoproterenol, but not in unstimulated cells, whereas serine-2808 was considerably phosphorylated before and after isoproterenol treatment. We further showed that serine-2030 was stoichiometrically phosphorylated by PKA, but not by CaMKII, and that mutations of serine-2030 altered neither the FKBP12.6-RyR2 interaction nor the Ca2+ dependence of [3H]ryanodine binding. Moreover, the levels of phosphorylation of RyR2 at serine-2030 and serine-2808 in both failing and non-failing canine hearts were similar. Together, our data indicate that serine-2030 is a major PKA phosphorylation site in RyR2 responding to acute beta-adrenergic stimulation, and that RyR2 is not hyperphosphorylated by PKA in canine HF.
- Published
- 2005
- Full Text
- View/download PDF
139. A highly sensitive method for quantification of myosin light chain phosphorylation by capillary isoelectric focusing with laser-induced fluorescence detection.
- Author
-
Shiraishi M, Loutzenhiser RD, and Walsh MP
- Subjects
- Animals, Chickens, Electrophoresis, Capillary methods, Electrophoresis, Polyacrylamide Gel methods, Gizzard, Avian chemistry, Isoelectric Focusing methods, Lasers, Phosphorylation, Sensitivity and Specificity, Spectrometry, Fluorescence, Spectrophotometry, Ultraviolet, Myosin Light Chains isolation & purification, Myosin Light Chains metabolism
- Abstract
Activation of myosin II by phosphorylation of the 20 kDa regulatory light chains (LC20) has been implicated in numerous contractile and motile events, e.g., smooth muscle contraction, cytokinesis, and cell migration. The ability to analyze LC20 phosphorylation in minute samples is critical to determine the importance of LC20 phosphorylation in diverse physiological processes. We have developed a method for the separation and quantification of unphosphorylated, monophosphorylated, and diphosphorylated LC20 with a detection limit of 1 pg (50 amol). LC20 is initially isolated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transblotted to a polyvinlyidene difluoride (PVDF) membrane. The region of the membrane containing the LC20 band (identified by electrophoresis of purified LC20 in a neighboring lane) is cut out and fluorescently labeled with Alexa Fluor 488 C5 maleimide. The labeled LC20 is eluted from the membrane with detergent and subjected to capillary isoelectric focusing (CIEF) to separate unphosphorylated, mono-, and diphosphorylated LC20, which are detected and quantified by laser-induced fluorescence (LIF). A linear relationship between log(peak area) and log(LC20 amount) is observed over the range of 50 amol-150 fmol. Quantification of LC20 phosphorylation by CIEF with LIF detection was compared with three commonly used methods with much lower levels of sensitivity: urea/glycerol-PAGE with Western blotting, phosphorylation by [gamma-32P]ATP with Cerenkov counting, and phosphorylation by [gamma-32P]ATP followed by SDS-PAGE, autoradiography, and scanning densitometry. All four methods gave very similar quantitative results, the major difference being that the new method exhibits 3000-fold enhanced sensitivity. This method is therefore applicable to quantitative analysis of phosphorylation of minute quantities of LC20.
- Published
- 2005
- Full Text
- View/download PDF
140. Identification of the linker histone H1 as a protein kinase Cepsilon-binding protein in vascular smooth muscle.
- Author
-
Zhao M, Sutherland C, Wilson DP, Deng J, Macdonald JA, and Walsh MP
- Subjects
- Amino Acid Sequence, Animals, Calcium metabolism, Diglycerides metabolism, Isoenzymes metabolism, Male, Molecular Sequence Data, Phosphorylation, Protein Binding, Protein Kinase C-epsilon, Rats, Rats, Sprague-Dawley, Receptors for Activated C Kinase, Receptors, Cell Surface metabolism, Aorta metabolism, Histones metabolism, Muscle, Smooth, Vascular metabolism, Phospholipids metabolism, Protein Kinase C metabolism
- Abstract
A variety of anchoring proteins target specific protein kinase C (PKC) isoenzymes to particular subcellular locations or multimeric signaling complexes, thereby achieving a high degree of substrate specificity by localizing the kinase in proximity to specific substrates. PKCepsilon is widely expressed in smooth muscle tissues, but little is known about its targeting and substrate specificity. We have used a Far-Western (overlay) approach to identify PKCepsilon-binding proteins in vascular smooth muscle of the rat aorta. Proteins of approximately 32 and 34 kDa in the Triton-insoluble fraction were found to bind PKCepsilon in a phospholipid/diacylglycerol-dependent manner. Although of similar molecular weight to RACK-1, a known PKCepsilon-binding protein, these proteins were separated from RACK-1 by SDS-PAGE and differential NaCl extraction and were not recognized by an antibody to RACK-1. The PKCepsilon-binding proteins were further purified from the Triton-insoluble fraction and identified by de novo sequencing of selected tryptic peptides by tandem mass spectrometry as variants of the linker histone H1. Their identity was confirmed by Western blotting with anti-histone H1 and the demonstration that purified histone H1 binds PKCepsilon in the presence of phospholipid and diacylglycerol but absence of Ca(2+). The interaction of PKCepsilon with histone H1 was specific since no interaction was observed with histones H2A, H2S or H3S. Bound PKCepsilon phosphorylated histone H1 in a phospholipid/diacylglycerol-dependent but Ca(2+)-independent manner. Ca(2+)-dependent PKC was also shown to interact with histone H1 but not other histones. These results suggest that histone H1 is both an anchoring protein and a substrate for activated PKCepsilon and other PKC isoenzymes and likely serves to localize activated PKCs that translocate to the nucleus in the vicinity of specific nuclear substrates including histone H1 itself. Since PKC isoenzymes have been implicated in regulation of gene expression, stable interaction with histone H1 may be an important step in this process.
- Published
- 2004
- Full Text
- View/download PDF
141. Distinct kinases are involved in contraction of cat esophageal and lower esophageal sphincter smooth muscles.
- Author
-
Kim N, Cao W, Song IS, Kim CY, Harnett KM, Cheng L, Walsh MP, and Biancani P
- Subjects
- Animals, Antibodies, Cats, Enzyme Inhibitors pharmacology, Female, Male, Marine Toxins, Microcystins, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinases metabolism, Muscle Contraction drug effects, Muscle, Smooth cytology, Myocytes, Smooth Muscle enzymology, Myosin-Light-Chain Kinase metabolism, Okadaic Acid pharmacology, Peptides, Cyclic pharmacology, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoprotein Phosphatases immunology, Phosphoprotein Phosphatases metabolism, Protein Kinase C-epsilon, Protein Phosphatase 1, Protein Serine-Threonine Kinases metabolism, Esophagogastric Junction enzymology, Mitogen-Activated Protein Kinase Kinases metabolism, Muscle Contraction physiology, Muscle, Smooth enzymology, Protein Kinase C metabolism
- Abstract
Contraction of smooth muscle depends on the balance of myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP) activities. Because MLCK activation depends on the activation of calmodulin, which requires a high Ca(2+) concentration, phosphatase inhibition has been invoked to explain contraction at low cytosolic Ca(2+) levels. The link between activation of the Ca(2+)-independent protein kinase Cepsilon (PKCepsilon) and MLC phosphorylation observed in the esophagus (ESO) (Sohn UD, Cao W, Tang DC, Stull JT, Haeberle JR, Wang CLA, Harnett KM, Behar J, and Biancani P. Am J Physiol Gastrointest Liver Physiol 281: G467-G478, 2001), however, has not been elucidated. We used phosphatase and kinase inhibitors and antibodies to signaling enzymes in combination with intact and saponin-permeabilized isolated smooth muscle cells from ESO and lower esophageal sphincter (LES) to examine PKCepsilon-dependent, Ca(2+)-independent signaling in ESO. The phosphatase inhibitors okadaic acid and microcystin-LR, as well as an antibody to the catalytic subunit of type 1 protein serine/threonine phosphatase, elicited similar contractions in ESO and LES. MLCK inhibitors (ML-7, ML-9, and SM-1) and antibodies to MLCK inhibited contraction induced by phosphatase inhibition in LES but not in ESO. The PKC inhibitor chelerythrine and antibodies to PKCepsilon, but not antibodies to PKCbetaII, inhibited contraction of ESO but not of LES. In ESO, okadaic acid triggered translocation of PKCepsilon from cytosolic to particulate fraction and increased activity of integrin-linked kinase (ILK). Antibodies to the mitogen-activated protein (MAP) kinases ERK1/ERK2 and to ILK, and the MAP kinase kinase (MEK) inhibitor PD-98059, inhibited okadaic acid-induced ILK activity and contraction of ESO. We conclude that phosphatase inhibition potentiates the effects of MLCK in LES but not in ESO. Contraction of ESO is mediated by activation of PKCepsilon, MEK, ERK1/2, and ILK.
- Published
- 2004
- Full Text
- View/download PDF
142. Protein kinase A phosphorylation at serine-2808 of the cardiac Ca2+-release channel (ryanodine receptor) does not dissociate 12.6-kDa FK506-binding protein (FKBP12.6).
- Author
-
Xiao B, Sutherland C, Walsh MP, and Chen SR
- Subjects
- Amino Acid Substitution, Animals, Antigen-Antibody Reactions, Calcium Signaling genetics, Cell Line, Dogs, Humans, Kidney, Macromolecular Substances, Mice, Myocytes, Cardiac drug effects, Phosphorylation, Phosphoserine chemistry, Protein Binding, Recombinant Fusion Proteins metabolism, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel genetics, Ryanodine Receptor Calcium Release Channel immunology, Sarcoplasmic Reticulum metabolism, Sirolimus pharmacology, Structure-Activity Relationship, Calcium Signaling physiology, Cyclic AMP-Dependent Protein Kinases metabolism, Mutation, Missense, Myocytes, Cardiac metabolism, Point Mutation, Protein Processing, Post-Translational, Ryanodine Receptor Calcium Release Channel metabolism, Tacrolimus Binding Proteins metabolism
- Abstract
Dissociation of FKBP12.6 from the cardiac Ca2+-release channel (RyR2) as a consequence of protein kinase A (PKA) hyperphosphorylation of RyR2 at a single amino acid residue, serine-2808, has been proposed as an important mechanism underlying cardiac dysfunction in heart failure. However, the issue of whether PKA phosphorylation of RyR2 can dissociate FKBP12.6 from RyR2 is controversial. To additionally address this issue, we investigated the effect of PKA phosphorylation and mutations at serine-2808 of RyR2 on recombinant or native FKBP12.6-RyR2 interaction. Site-specific antibodies, which recognize the serine-2808 phosphorylated or nonphosphorylated form of RyR2, were used to unambiguously correlate the phosphorylation state of RyR2 at serine-2808 with its ability to bind FKBP12.6. We found that FKBP12.6 can bind to both the serine-2808 phosphorylated and nonphosphorylated forms of RyR2. The S2808D mutant thought to mimic constitutive phosphorylation also retained the ability to bind FKBP12.6. Complete phosphorylation at serine-2808 by exogenous PKA disrupted neither the recombinant nor native FKBP12.6-RyR2 complex. Furthermore, binding of site-specific antibodies to the serine-2808 phosphorylation site did not dissociate FKBP12.6 from or prevent FKBP12.6 from binding to RyR2. Taken together, our results do not support the notion that PKA phosphorylation at serine-2808 dissociates FKBP12.6 from RyR2.
- Published
- 2004
- Full Text
- View/download PDF
143. Myosin heavy chain expression in renal afferent and efferent arterioles: relationship to contractile kinetics and function.
- Author
-
Shiraishi M, Wang X, Walsh MP, Kargacin G, Loutzenhiser K, and Loutzenhiser R
- Subjects
- Angiotensin II pharmacology, Animals, Arterioles drug effects, Arterioles metabolism, Arterioles physiology, Kidney Glomerulus blood supply, Kinetics, Myosin Heavy Chains genetics, Norepinephrine pharmacology, Protein Isoforms genetics, Protein Isoforms metabolism, RNA, Messenger metabolism, Renal Artery drug effects, Transcription, Genetic, Myosin Heavy Chains metabolism, Renal Artery metabolism, Renal Artery physiology, Vasoconstriction
- Abstract
The physiological role of smooth muscle myosin heavy chain (MHC) isoform diversity is poorly understood. The expression of MHC-B, which contains an insert at the ATP binding pocket, has been linked to enhanced contractile kinetics. We recently reported that the renal afferent arteriole exhibits an unusually rapid myogenic response and that its kinetic features allow this vessel to modulate tone in response to alterations in systolic blood pressure. In the present study, we examined MHC expression patterns in renal afferent and efferent arterioles. These two vessels regulate glomerular inflow and outflow resistances and control the pressure within the intervening glomerular capillaries (PGC). Whereas the afferent arteriole must respond rapidly to increases in blood pressure, the efferent arteriole plays a distinctly different role, maintaining a tonic elevation in outflow resistance to preserve function when renal perfusion is compromised. Using RT-PCR, Western analysis, and immunofluorescence imaging of intact isolated arterioles, we found that the afferent arteriole predominantly expresses the MHC-B isoform, whereas the efferent arteriole expresses only the slower-cycling MHC-A isoform. We examined the kinetics of angiotensin II- and norepinephrine-induced vasoconstriction and found that the afferent arteriole responds approximately 3-fold faster than the efferent arteriole. Our findings thus point to the renal microcirculation as a unique and important example of smooth muscle adaptation in regard to MHC isoform expression and physiological function.
- Published
- 2003
- Full Text
- View/download PDF
144. Unmasking the annexin I interaction from the structure of Apo-S100A11.
- Author
-
Dempsey AC, Walsh MP, and Shaw GS
- Subjects
- Annexin A1 chemistry, Dimerization, Nuclear Magnetic Resonance, Biomolecular, S100 Proteins chemistry, Annexin A1 metabolism, S100 Proteins metabolism
- Abstract
S100A11 is a homodimeric EF-hand calcium binding protein that undergoes a calcium-induced conformational change and interacts with the phospholipid binding protein annexin I to coordinate membrane association. In this work, the solution structure of apo-S100A11 has been determined by NMR spectroscopy to uncover the details of its calcium-induced structural change. Apo-S100A11 forms a tight globular structure having a near antiparallel orientation of helices III and IV in calcium binding site II. Further, helices I and IV, and I and I', form a more closed arrangement than observed in other apo-S100 proteins. This helix arrangement in apo-S100A11 partially buries residues in helices I (P3, E11, A15), III (V55, R58, M59), and IV (A86, C87, S90) and the linker (A45, F46), which are required for interaction with annexin I in the calcium-bound state. In apo-S100A11, this results in a "masked" binding surface that prevents annexin I binding but is uncovered upon calcium binding.
- Published
- 2003
- Full Text
- View/download PDF
145. Phosphorylation of protein phosphatase type-1 inhibitory proteins by integrin-linked kinase and cyclic nucleotide-dependent protein kinases.
- Author
-
Erdodi F, Kiss E, Walsh MP, Stefansson B, Deng JT, Eto M, Brautigan DL, and Hartshorne DJ
- Subjects
- Animals, Chickens, Cyclic AMP-Dependent Protein Kinases metabolism, Dose-Response Relationship, Drug, Humans, Inhibitory Concentration 50, Intracellular Signaling Peptides and Proteins, Muscle Proteins metabolism, Mutation, Myosin-Light-Chain Phosphatase, Phosphoproteins metabolism, Phosphoric Monoester Hydrolases metabolism, Protein Phosphatase 1, Proteins metabolism, Time Factors, Turkeys, Cyclin-Dependent Kinases metabolism, Phosphoprotein Phosphatases metabolism, Phosphorylation, Protein Serine-Threonine Kinases metabolism
- Abstract
Protein phosphatases play key roles in cellular regulation and are subjected to control by protein inhibitors whose activity is in turn regulated by phosphorylation. Here we investigated the possible regulation of phosphorylation-dependent type-1 protein phosphatase (PP1) inhibitors, CPI-17, PHI-1, and KEPI, by various kinases. Protein kinases A (PKA) and G (PKG) phosphorylated CPI-17 at the inhibitory site (T38), but not PHI-1 (T57). Phosphorylated CPI-17 inhibited the activity of both the PP1 catalytic subunit (PP1c) and the myosin phosphatase holoenzyme (MPH) with IC(50) values of 1-8 nM. PKA predominantly phosphorylated a site distinct from the inhibitory T73 in KEPI, whereas PKG was ineffective. Integrin-linked kinase phosphorylated KEPI (T73) and this dramatically increased inhibition of PP1c (IC(50)=0.1 nM) and MPH (IC(50)=8 nM). These results suggest that the regulatory phosphorylation of CPI-17 and KEPI may involve distinct kinases and signaling pathways.
- Published
- 2003
- Full Text
- View/download PDF
146. The role of RhoA and Rho-associated kinase in vascular smooth muscle contraction.
- Author
-
Swärd K, Mita M, Wilson DP, Deng JT, Susnjar M, and Walsh MP
- Subjects
- Animals, Calcium physiology, Humans, Hypertension metabolism, Intracellular Signaling Peptides and Proteins, Myosin-Light-Chain Phosphatase, Phosphoprotein Phosphatases metabolism, Phosphorylation, Signal Transduction, rho-Associated Kinases, Hypertension physiopathology, Muscle Contraction physiology, Muscle, Smooth, Vascular physiology, Protein Serine-Threonine Kinases physiology, rhoA GTP-Binding Protein physiology
- Abstract
A variety of contractile agonists trigger activation of the small GTPase RhoA. An important target of activated RhoA in smooth muscle is Rho-associated kinase (ROK), one of the downstream targets that is the myosin binding subunit (MYPT1) of myosin light chain phosphatase (MLCP). Phosphorylation of MYPT1 at T695 by activated ROK results in a decrease in phosphatase activity of MLCP and an increase in myosin light chain (LC(20)) phosphorylation catalyzed by Ca(2)(+)/calmodulin-dependent myosin light chain kinase and/or a distinct Ca(2)(+)-independent kinase. LC(20) phosphorylation in turn triggers cross-bridge cycling and force development. ROK also phosphorylates the cytosolic protein CPI-17 (at T38), which thereby becomes a potent inhibitor of MLCP. The RhoA/ROK pathway has been implicated in the tonic phase of force maintenance in response to various agonists, with no evident role in the phasic response, suggesting this pathway as a potential target for antihypertensive therapy. Indeed, ROK inhibitors restore normal blood pressure in several rat hypertensive models.
- Published
- 2003
- Full Text
- View/download PDF
147. Regulation of volume-sensitive outwardly rectifying anion channels in pulmonary arterial smooth muscle cells by PKC.
- Author
-
Zhong J, Wang GX, Hatton WJ, Yamboliev IA, Walsh MP, and Hume JR
- Subjects
- Animals, Biological Transport drug effects, Dogs, Enzyme Inhibitors pharmacology, Isoenzymes metabolism, Muscle, Smooth, Vascular drug effects, Myocytes, Smooth Muscle cytology, Myocytes, Smooth Muscle drug effects, Phorbol 12,13-Dibutyrate pharmacology, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Protein Kinase C-epsilon, Pulmonary Artery cytology, Pulmonary Artery drug effects, Tissue Distribution, Anions metabolism, Ion Channels metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Protein Kinase C physiology, Pulmonary Artery metabolism
- Abstract
We tested the possible role of endogenous protein kinase C (PKC) in the regulation of native volume-sensitive organic osmolyte and anion channels (VSOACs) in acutely dispersed canine pulmonary artery smooth muscle cells (PASMC). Hypotonic cell swelling activated native volume-regulated Cl(-) currents (I(Cl.vol)) which could be reversed by exposure to phorbol 12,13-dibutyrate (0.1 microM) or by hypertonic cell shrinkage. Under isotonic conditions, calphostin C (0.1 microM) or Ro-31-8425 (0.1 microM), inhibitors of both conventional and novel PKC isozymes, significantly activated I(Cl.vol) and prevented further modulation by subsequent hypotonic cell swelling. Bisindolylmaleimide (0.1 microM), a selective conventional PKC inhibitor, was without effect. Dialyzing acutely dispersed and cultured PASMC with epsilon V1-2 (10 microM), a translocation inhibitory peptide derived from the V1 region of epsilon PKC, activated I(Cl.vol) under isotonic conditions and prevented further modulation by cell volume changes. Dialyzing PASMC with beta C2-2 (10 microM), a translocation inhibitory peptide derived from the C2 region of beta PKC, had no detectable effect. Immunohistochemistry in cultured canine PASMC verified that hypotonic cell swelling is accompanied by translocation of epsilon PKC from the vicinity of the membrane to cytoplasmic and perinuclear locations. These data suggest that membrane-bound epsilon PKC controls the activation state of native VSOACs in canine PASMC under isotonic and anisotonic conditions.
- Published
- 2002
- Full Text
- View/download PDF
148. Phosphorylation of the myosin phosphatase inhibitors, CPI-17 and PHI-1, by integrin-linked kinase.
- Author
-
Deng JT, Sutherland C, Brautigan DL, Eto M, and Walsh MP
- Subjects
- Animals, Calcium metabolism, In Vitro Techniques, Male, Muscle Contraction drug effects, Muscle Proteins genetics, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Myosin-Light-Chain Phosphatase, Octoxynol pharmacology, Peptide Mapping, Phosphoprotein Phosphatases metabolism, Phosphoproteins genetics, Phosphorylation, Point Mutation, Proteins genetics, Rats, Rats, Sprague-Dawley, Muscle Proteins metabolism, Phosphoprotein Phosphatases antagonists & inhibitors, Phosphoproteins metabolism, Protein Serine-Threonine Kinases metabolism, Proteins metabolism
- Abstract
Integrin-linked kinase (ILK) has been implicated in Ca(2+)- independent contraction of smooth muscle via its ability to phosphorylate myosin. We investigated the possibility that this kinase might also phosphorylate and regulate the myosin light-chain phosphatase inhibitor proteins CPI-17 [protein kinase C (PKC)-dependent phosphatase inhibitor of 17 kDa] and PHI-1 (phosphatase holoenzyme inhibitor-1), known substrates of PKC. Both phosphatase inhibitors were phosphorylated by ILK in an in-gel kinase assay and in solution. A Thr-->Ala mutation at Thr(38) of CPI-17 and Thr(57) of PHI-1 eliminated phosphorylation by ILK. Phosphopeptide mapping, phospho amino acid analysis and immunoblotting using phospho-specific antibodies indicated that ILK predominantly phosphorylated the site critical for potent inhibition, i.e. Thr(38) of CPI-17 or Thr(57) of PHI-1. CPI-17 and PHI-1 thiophosphorylated by ILK at Thr(38) or Thr(57) respectively inhibited myosin light-chain phosphatase (MLCP) activity bound to myosin, whereas the site-specific mutants CPI-17-Thr(38)Ala and PHI-1-Thr(57)Ala, treated with ILK under identical conditions, like the untreated wild-type proteins had no effect on the phosphatase. Consistent with these effects, both thiophospho-CPI-17 and -PHI-1 induced Ca(2+) sensitization of contraction of Triton X-100-demembranated rat-tail arterial smooth muscle, whereas CPI-17-Thr(38)Ala and PHI-1-Thr(57)Ala treated with ILK in the presence of adenosine 5'-[gamma-thio]triphosphate failed to evoke a contractile response. We conclude that ILK may activate smooth-muscle contraction both directly, via phosphorylation of myosin, and indirectly, via phosphorylation and activation of CPI-17 and PHI-1, leading to inhibition of MLCP.
- Published
- 2002
- Full Text
- View/download PDF
149. Quantum dot superlattice thermoelectric materials and devices.
- Author
-
Harman TC, Taylor PJ, Walsh MP, and LaForge BE
- Abstract
PbSeTe-based quantum dot superlattice structures grown by molecular beam epitaxy have been investigated for applications in thermoelectrics. We demonstrate improved cooling values relative to the conventional bulk (Bi,Sb)2(Se,Te)3 thermoelectric materials using a n-type film in a one-leg thermoelectric device test setup, which cooled the cold junction 43.7 K below the room temperature hot junction temperature of 299.7 K. The typical device consists of a substrate-free, bulk-like (typically 0.1 millimeter in thickness, 10 millimeters in width, and 5 millimeters in length) slab of nanostructured PbSeTe/PbTe as the n-type leg and a metal wire as the p-type leg.
- Published
- 2002
- Full Text
- View/download PDF
150. Phosphorylation of the myosin phosphatase target subunit by integrin-linked kinase.
- Author
-
Murányi A, MacDonald JA, Deng JT, Wilson DP, Haystead TA, Walsh MP, Erdodi F, Kiss E, Wu Y, and Hartshorne DJ
- Subjects
- Amino Acid Sequence, Animals, Blotting, Western, Catalytic Domain, Chickens, Chromatography, High Pressure Liquid, Cyclic AMP metabolism, Dose-Response Relationship, Drug, Molecular Sequence Data, Muscle, Smooth cytology, Mutation, Myosin-Light-Chain Phosphatase, Phosphorylation, Protein Binding, Protein Structure, Tertiary, Protein Transport, Serine metabolism, Threonine metabolism, Time Factors, Phosphoprotein Phosphatases metabolism, Protein Serine-Threonine Kinases chemistry
- Abstract
A mechanism proposed for regulation of myosin phosphatase (MP) activity is phosphorylation of the myosin phosphatase target subunit (MYPT1). Integrin-linked kinase (ILK) is associated with the contractile machinery and can phosphorylate myosin at the myosin light-chain kinase sites. The possibility that ILK may also phosphorylate and regulate MP was investigated. ILK was associated with the MP holoenzyme, shown by Western blots and in-gel kinase assays. MYPT1 was phosphorylated by ILK and phosphorylation sites in the N- and C-terminal fragments of MYPT1 were detected. From sequence analyses, three sites were identified: a primary site at Thr(709), and two other sites at Thr(695) and Thr(495). One of the sites for cAMP-dependent protein kinase (PKA) was Ser(694). Assays with the catalytic subunit of type 1 phosphatase indicated that only the C-terminal fragment of MYPT1 phosphorylated by zipper-interacting protein kinase, and ILK inhibited activity. The phosphorylated N-terminal fragment activated phosphatase activity and phosphorylation by PKA was without effect. Using full-length MYPT1 constructs phosphorylated by various kinases it was shown that Rho kinase gave marked inhibition; ILK produced an intermediate level of inhibition, which was considerably reduced for the Thr(695)-->Ala mutant; and PKA had no effect. In summary, phosphorylation of the various sites indicated that Thr(695) was the major inhibitory site, Thr(709) had only a slight inhibitory effect and Ser(694) had no effect. The findings that ILK phosphorylated both MYPT1 and myosin and the association of ILK with MP suggest that ILK may influence cytoskeletal structure or function.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.