Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Smooth Muscle Res ; 58(0): 22-33, 2022.
Article in English | MEDLINE | ID: mdl-35418530

ABSTRACT

CPI-17 regulates the myosin phosphatase and mediates the agonist-induced contraction of smooth muscle. PKC and ROCK phosphorylate CPI-17 at Thr38 leading to a conformational change of the central inhibitory domain (PHIN domain). The N- and C-terminal tails of CPI-17 are predicted as unstructured loops and their sequences are conserved among mammals. Here we characterized CPI-17 N- and C-terminal unstructured tails using recombinant proteins that lack the potions. Recombinant CPI-17 proteins at a physiologic level (10 µM) were doped into beta-escin-permeabilized smooth muscle strips for Ca2+ sensitization force measurement. The ectopic full-length CPI-17 augmented the PDBu-induced Ca2+ sensitization force at pCa6.3, indicating myosin phosphatase inhibition. Deletion of N- and C-terminal tails of CPI-17 attenuated the extent of PDBu-induced Ca2+-sensitization force. The N-terminal deletion dampened phosphorylation at Thr38 by protein kinase C (PKC), and the C-terminal truncation lowered the affinity to the myosin phosphatase. Under the physiologic conditions, PKC and myosin phosphatase may recognize CPI-17 N-/C-terminal unstructured tails inducing Ca2+ sensitization force in smooth muscle cells.


Subject(s)
Muscle Contraction , Muscle Proteins , Animals , Calcium/metabolism , Mammals/metabolism , Muscle Contraction/physiology , Muscle Proteins/metabolism , Muscle, Smooth/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C/metabolism
2.
Am J Physiol Cell Physiol ; 321(3): C549-C558, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34106787

ABSTRACT

Cell signaling pathways regulating myosin regulatory light chain (LC20) phosphorylation contribute to determining contractile responses in smooth muscles. Following excitation and contraction, phasic smooth muscles, such as the digestive tract and urinary bladder, undergo relaxation due to a decline of cellular Ca2+ concentration and decreased Ca2+ sensitivity of LC20 phosphorylation, named Ca2+ desensitization. Here, we determined the mechanisms underlying the temporal Ca2+ desensitization of LC20 phosphorylation in phasic smooth muscles using permeabilized strips of the mouse ileum and urinary bladder. Upon stimulation with pCa6.0 at 20°C, contraction and LC20 phosphorylation peaked within 30 s and then declined to about 50% of the peak force at 2 min after stimulation. During the relaxation phase after the contraction, LC20 kinase [myosin light chain kinase (MLCK)] was inactivated, but no fluctuation in LC20 phosphatase activity occurred, suggesting that MLCK inactivation is a cause of the Ca2+-induced Ca2+ desensitization of LC20 phosphorylation. MLCK inactivation was associated with phosphorylation at the calmodulin-binding domain of the kinase. Treatment with STO-609 and TIM-063 antagonists for Ca2+/calmodulin (CaM)-dependent protein kinase kinase-ß (CaMKKß) attenuated both the phasic response of the contraction and MLCK phosphorylation, whereas neither CaM kinase II, AMP-activated protein kinase, nor p21-activated kinase induced MLCK inactivation in phasic smooth muscles. Conversely, protein phosphatase 2A inhibition amplified the phasic response. Signaling pathways through CaMKKß and protein phosphatase 2A may contribute to regulating the phasic response of smooth muscle contraction.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/genetics , Myosin-Light-Chain Kinase/genetics , Protein Phosphatase 2/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Benzimidazoles/pharmacology , Calcium/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Female , Gene Expression Regulation , Ileum/metabolism , Mice , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Naphthalimides/pharmacology , Phosphorylation , Protein Phosphatase 2/metabolism , Tissue Culture Techniques , Urinary Bladder/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
3.
J Cell Physiol ; 233(1): 434-446, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28295256

ABSTRACT

Intercellular communication among autonomic nerves, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs) plays a central role in an uninterrupted regulation of blood flow through vascular contractile machinery. Impairment of this communication is linked to development of vascular diseases such as hypertension, cerebral/coronary vasospasms, aortic aneurism, and erectile dysfunction. Although the basic concept of the communication as a whole has been studied, the spatiotemporal correlation of ECs/VSMCs in tissues at the cellular level is unknown. Here, we show a unique VSMC response to ECs during contraction and relaxation of isolated aorta tissues through visualization of spatiotemporal activation patterns of smooth muscle myosin II. ECs in the intimal layer dictate the stimulus-specific heterogeneous activation pattern of myosin II in VSMCs within distinct medial layers. Myosin light chain (MLC) phosphorylation (active form of myosin II) gradually increases towards outer layers (approximately threefold higher MLC phosphorylation at the outermost layer than that of the innermost layer), presumably by release of an intercellular messenger, nitric oxide (NO). Our study also demonstrates that the MLC phosphorylation at the outermost layer in spontaneously hypertensive rats (SHR) during NO-induced relaxation is quite high and approximately 10-fold higher than that of its counterpart, the Wister-Kyoto rats (WKY), suggesting that the distinct pattern of myosin II activation within tissues is important for vascular protection against elevated blood pressure.


Subject(s)
Endothelial Cells/physiology , Fluorescent Antibody Technique , Microscopy, Fluorescence , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Vasoconstriction , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiology , Biomarkers , Cell Communication , Disease Models, Animal , Endothelial Cells/metabolism , Hypertension/metabolism , Hypertension/physiopathology , In Vitro Techniques , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Nitric Oxide/metabolism , Phosphorylation , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors , Vasodilation
4.
Pflugers Arch ; 469(12): 1651-1662, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28717991

ABSTRACT

Prolonged bed rest (PBR) causes orthostatic hypotension (OH). Rapid constriction of splanchnic resistance arteries in response to a sudden increase in sympathetic tone contributes to the recovery of orthostatic arterial pressure upon standing. However, the molecular mechanism of PBR-induced dysfunction in arterial constriction is not fully understood. Previously, we showed that CPI-17, a regulatory protein for myosin phosphatase, mediates α1A-adrenergic receptor-induced rapid contraction of small mesenteric arteries. Here, we tested whether PBR associated with OH affects the α1-adrenergic receptor-induced CPI-17 signaling pathway in mesenteric arteries using rats treated by head-down tail-suspension hindlimb unloading (HDU), an experimental OH model. In normal anesthetized rats, mean arterial pressure (MAP) rapidly reduced upon 90° head-up tilt from supine position and then immediately recovered without change in heart rate, suggesting a rapid arterial constriction. On the other hand, after a 4-week HDU treatment, the fast orthostatic MAP recovery failed for 1 min. Alpha1A subtype-specific antagonist suppressed the orthostatic MAP recovery with a small decrease in basal blood pressure, whereas non-specific α1-antagonist prazosin strongly reduced both basal MAP and orthostatic recovery. The HDU treatment resulted in 68% reduction in contraction in parallel with 83% reduction in CPI-17 phosphorylation in denuded mesenteric arteries 10 s after α1-agonist stimulation. The treatment with either Ca2+-release channel opener or PKC inhibitor mimicked the deficiency in HDU arteries. These results suggest that an impairment of the rapid PKC/CPI-17 signaling pathway downstream of α1A-adrenoceptors in peripheral arterial constriction, as an end organ of orthostatic blood pressure reflex, is associated with OH in prolonged bed rest patients.


Subject(s)
Bed Rest/adverse effects , Hypotension, Orthostatic/metabolism , Mesenteric Arteries/metabolism , Muscle Proteins/metabolism , Phosphoproteins/metabolism , Animals , Arterial Pressure/physiology , Female , Head-Down Tilt/adverse effects , Head-Down Tilt/physiology , Heart Rate/physiology , Hypotension, Orthostatic/etiology , Male , Phosphorylation , Rats , Rats, Sprague-Dawley
5.
J Smooth Muscle Res ; 53(0): 1-19, 2017.
Article in English | MEDLINE | ID: mdl-28260704

ABSTRACT

A hallmark of smooth muscle cells is their ability to adapt their functions to meet temporal and chronic fluctuations in their demands. These functions include force development and growth. Understanding the mechanisms underlying the functional plasticity of smooth muscles, the major constituent of organ walls, is fundamental to elucidating pathophysiological rationales of failures of organ functions. Also, the knowledge is expected to facilitate devising innovative strategies that more precisely monitor and normalize organ functions by targeting individual smooth muscles. Evidence has established a current paradigm that the myosin light chain phosphatase (MLCP) is a master regulator of smooth muscle responsiveness to stimuli. Cellular MLCP activity is negatively and positively regulated in response to G-protein activation and cAMP/cGMP production, respectively, through the MYPT1 regulatory subunit and an endogenous inhibitor protein named CPI-17. In this article we review the outcomes from two decade of research on the CPI-17 signaling and discuss emerging paradoxes in the view of signaling pathways regulating smooth muscle functions through MLCP.


Subject(s)
Muscle Contraction/genetics , Muscle, Smooth/physiology , Myosin-Light-Chain Phosphatase/physiology , Signal Transduction/physiology , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , GTP-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Muscle Proteins , Muscle Tonus/genetics , Myosin-Light-Chain Phosphatase/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/physiology , Phosphorylation , Signal Transduction/genetics , rho-Associated Kinases/physiology
6.
Prostate ; 73(9): 996-1006, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23389830

ABSTRACT

BACKGROUND: Prostate smooth muscle plays an important role in the physiological ejection of prostatic fluid and also in the pathogenesis of benign prostate hyperplasia. Although mouse is the best genetically engineered animal model to identify potential molecular targets for human diseases, only fragmentary information is available for basic mechanism of mouse prostate contraction. METHODS: Small smooth muscle tubular rings were excised from four mouse prostate lobes to measure their isometric contractions. High K(+) , noradrenaline (NA), or acetylcholine (ACh) was applied with and without various antagonists and/or inhibitors to examine the contractile signaling pathways. RESULTS: Maximum amplitude of agonist-induced contractions varied greatly with different lobes but not with different locations or orientations within each lobe. Both NA and ACh produced large contractions in ventral and dorsal rings, whereas only small contractions were elicited in lateral and anterior rings. Combination of alpha-1 and muscarinic antagonists suppressed K(+) depolarization-induced contraction potently in ventral rings, but slightly in anterior rings. Blocking of either Ca(2+) -release or Ca(2+) -influx reduced agonist-induced contraction of ventral rings, however, a considerable amount of contractility remained even with both blockers. Inhibitors of ROCK and PKC partially inhibited NA-induced contractions, whereas a combination of Ca(2+) -blockers and Ca(2+) -sensitization inhibitors strongly suppressed the contraction. CONCLUSIONS: The ejection of prostatic fluid is differentially regulated in each prostate lobe. In ventral prostate smooth muscle, Ca(2+) -release, Ca(2+) -influx, and ROCK- and PKC-mediated Ca(2+) -sensitizations are all involved in NA-induced contractions. This finding is a useful step toward the understanding of the phenotypic changes in the smooth muscle of BPH prostate.


Subject(s)
Muscle, Smooth/physiology , Prostate/physiology , Acetylcholine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Atropine/pharmacology , Calcium/pharmacology , Calcium Channel Blockers/pharmacology , Cholinergic Agonists/pharmacology , In Vitro Techniques , Male , Mice , Muscarinic Antagonists/pharmacology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/anatomy & histology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Nicardipine/pharmacology , Norepinephrine/pharmacology , Prazosin/pharmacology , Prostate/anatomy & histology , Prostate/drug effects , Prostate/metabolism , Protein Kinase Inhibitors/pharmacology , Ryanodine/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
7.
Pflugers Arch ; 465(2): 283-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23149489

ABSTRACT

Smooth muscle caldesmon (h-CaD) is an actin- and myosin-binding protein that reversibly inhibits the actomyosin ATPase activity in vitro. To test the function of h-CaD in vivo, we eliminated its expression in mice. The h-CaD-null animals appeared normal and fertile, although the litter size was smaller. Tissues from the homozygotes lacked h-CaD and exhibited upregulation of the non-muscle isoform, l-CaD, in visceral, but not vascular tonic smooth muscles. While the Ca(2+) sensitivity of force generation of h-CaD-deficient smooth muscle remained largely unchanged, the kinetic behavior during relaxation in arteries was different. Both intact and permeabilized arterial smooth muscle tissues from the knockout animals relaxed more slowly than those of the wild type. Since this difference occurred after myosin dephosphorylation was complete, the kinetic effect most likely resulted from slower detachment of unphosphorylated crossbridges. Detailed analyses revealed that the apparently slower relaxation of h-CaD-null smooth muscle was due to an increase in the amplitude of a slower component of the biphasic tension decay. While the identity of this slower process has not been unequivocally determined, we propose it reflects a thin filament state that elicits fewer re-attached crossbridges. Our finding that h-CaD modulates the rate of smooth muscle relaxation clearly supports a role in the control of vascular tone.


Subject(s)
Calmodulin-Binding Proteins/genetics , Muscle Relaxation/genetics , Muscle, Smooth, Vascular/metabolism , Animals , Arteries/metabolism , Arteries/pathology , Arteries/physiopathology , Calmodulin-Binding Proteins/metabolism , Homozygote , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myosins/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Up-Regulation , Vasodilation/genetics
8.
Int J Clin Exp Pathol ; 5(8): 787-95, 2012.
Article in English | MEDLINE | ID: mdl-23071861

ABSTRACT

During the influenza pandemic of 2009, the number of viral pneumonia cases showed a marked increase in comparison with seasonal influenza viruses. Mutations at amino acid 222 (D222G mutations) in the virus hemagglutinin (HA) molecule, known to alter the receptor-recognition properties of the virus, were detected in a number of the more severely-affected patients in the early phases of the pandemic. To understand the background for the emergence of the mutant amino acid D222G in human lungs, we conducted histological examinations on lung specimens of patients from Mexico who had succumbed in the pandemic. Prominent regenerative and hyperplastic changes in the alveolar type II pneumocytes, which express avian-type sialoglycan receptors in the respiratory tract of severely affected individuals, were observed in the Mexican patients. An infection model utilizing guinea pigs, which was chosen in order to best simulate the sialic acid distribution of severe pneumonia in human patients, demonstrated an increase of D222G mutants and a delay in the diminution of mutants in the lower respiratory tract in comparison to the upper respiratory tract. Our data suggests that the predominance of avian-type sialoglycan receptors in the pneumonic lungs may contribute to the emergence of viral HA mutants. This data comprehensively illustrates the mechanisms for the emergence of mutants in the clinical samples.


Subject(s)
Disease Models, Animal , Hemagglutinins, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/virology , Orthomyxoviridae Infections/virology , Pneumonia, Viral/virology , Alveolar Epithelial Cells/virology , Animals , Disease Outbreaks , Female , Genes, Viral , Guinea Pigs , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/pathology , Madin Darby Canine Kidney Cells , Mutation , Pneumonia, Viral/pathology , RNA, Viral/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
J Physiol ; 590(21): 5401-23, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22930267

ABSTRACT

Each segment along arterial vessels adapts to different circumstances, including blood pressure and sympathetic innervation. PKC and Rho-associated kinase (ROCK) Ca(2+)-sensitizing pathways leading to myosin phosphatase inhibition are critically involved in α(1)-adrenoceptor-mediated vascular smooth muscle contraction in distinctive time-dependent manners. We tested whether the amplitude and time course of each pathway varies dynamically between arterial segments. Using pharmacological approaches, we determined the time-dependent roles of Ca(2+) release, Ca(2+) influx, PKC and ROCK in α(1)-agonist-induced contraction and phosphorylation of key proteins in denuded rat small mesenteric artery, midsized caudal artery and thoracic aorta. SR Ca(2+) release and voltage-dependent Ca(2+) influx were essential for the initial rising and late sustained phases, respectively, of phenylephrine-induced contraction, regardless of arterial size. In small mesenteric arteries, α(1A)-subtype-specific antagonists and inhibitors of PKC, but not ROCK, markedly reduced the initial and late phases of contraction in a non-additive manner and suppressed phosphorylation of myosin light chain (MLC) and CPI-17, but not myosin targeting subunit of myosin light chain phosphatase (MYPT1). In aorta, an α(1D)-specific antagonist reduced both the initial and late phases of contraction with a significant decrease in MLC but not CPI-17 or MYPT1 phosphorylation. ROCK inhibitors, but not PKC inhibitors, suppressed the sustained phase of contraction with a decrease in MLC and MYPT1 phosphorylation in the aorta. The effect of ROCK inhibitors was additive with the α(1D)-antagonist. The results for midsized arteries were intermediate. Thus, the PKC-CPI-17 Ca(2+)-sensitizing pathway, which is dependent on PKC subtype and a Ca(2+)-handling mechanism, and is downstream of α(1A) receptors, plays a major role in α(1)-agonist-induced contraction of small resistance arteries in the splanchnic vascular beds. The effect of PKC and ROCK increases and decreases, respectively, with decreasing arterial size.


Subject(s)
Arteries/physiology , Calcium/physiology , Muscle, Smooth, Vascular/physiology , Protein Kinase C/physiology , Receptors, Adrenergic, alpha-1/physiology , rho-Associated Kinases/physiology , Amides/pharmacology , Animals , Arteries/drug effects , Enzyme Inhibitors/pharmacology , Female , In Vitro Techniques , Male , Muscle, Smooth, Vascular/drug effects , Protein Kinase C/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vasoconstriction/drug effects , rho-Associated Kinases/antagonists & inhibitors
10.
J Muscle Res Cell Motil ; 32(2): 65-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21643972

ABSTRACT

Chronic treatment with fetal bovine serum (FBS) causes contractility reduction, morphological alteration and DNA synthesis in organ-cultured vascular tissues. Here, we tested the hypothesis that chronic inhibition of ROCK has a protective effect on FBS-induced alterations in small arteries. Rabbit mesenteric arterial rings were cultured in FBS-supplemented culture medium with or without Y-27632, a reversible ROCK inhibitor. Chronic Y-27632 treatment prevented FBS-induced gradual arterial constriction, wall thickening, reduced contractility, and increased ROCK-specific MYPT1 Thr853 phosphorylation. Treatment with Y-27632 also prevented decreased eNOS mRNA expression, and reduced acetylcholine-induced relaxation. Sudden application of Y-27632 to pre-cultured rings reduced MYPT1 phosphorylation and re-widened the constricted rings. Chronic treatment with Y-27632, however, rather augmented than reduced the FBS-induced RhoA over-expression, also increased ROCK1 and MYPT1 expression and averted the FBS-induced reduction of MLC expression, suggesting a compensation of inhibited RhoA/ROCK activity. Sudden removal of Y-27632 caused a rebound in MYPT1 phosphorylation and vasoconstriction in rabbit mesenteric artery. To test which ROCK isoform has greater involvement in FBS-induced contraction, haploinsufficient Rock1+/- and Rock2+/- mouse mesenteric arterial rings were subjected to organ-culture. FBS-induced contraction and RhoA over-expression in either heterozygous animal was not different from wild-type animals. These results suggest that FBS-induced contraction is mediated by up-regulation of RhoA and subsequent activation of ROCK. In conclusion, chronic ROCK inhibition produces some effects that protect against FBS-stimulated vasoconstriction and remodeling. There are also negative effects that a sudden withdrawal of ROCK inhibitor might cause a stronger vasoconstriction than before it was used.


Subject(s)
Amides/pharmacology , Enzyme Inhibitors/pharmacology , Mesenteric Arteries/enzymology , Muscle, Smooth, Vascular/enzymology , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Animals , Cattle , Constriction, Pathologic/enzymology , Constriction, Pathologic/pathology , Mesenteric Arteries/pathology , Mice , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/pathology , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase , Organ Culture Techniques , Phosphorylation/drug effects , Rabbits , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
11.
PLoS One ; 6(6): e21207, 2011.
Article in English | MEDLINE | ID: mdl-21701593

ABSTRACT

Avian influenza H5N1 and pandemic (H1N1) 2009 viruses are known to induce viral pneumonia and subsequent acute respiratory distress syndrome (ARDS) with diffuse alveolar damage (DAD). The mortality rate of ARDS/DAD is extremely high, at approximately 60%, and no effective treatment for ARDS/DAD has been established. We examined serial pathological changes in the lungs of mice infected with influenza virus to determine the progress from viral pneumonia to ARDS/DAD. Mice were intranasally infected with influenza A/Puerto Rico/8/34 (PR8) virus, and their lungs were examined both macro- and micro-pathologically every 2 days. We also evaluated general condition, survival rate, body weight, viral loads in lung, and surfactant proteins in serum. As a result, all infected mice died within 9 days postinfection. At 2 days postinfection, inflammation in alveolar septa, i.e., interstitial pneumonia, was observed around bronchioles. From 4 to 6 days postinfection, interstitial pneumonia with alveolar collapse expanded throughout the lungs. From 6 to 9 days postinfection, DAD with severe alveolar collapse was observed in the lungs of all of dying and dead mice. In contrast, DAD was not observed in the live infected-mice from 2 to 6 days postinfection, despite their poor general condition. In addition, histopathological analysis was performed in mice infected with a dose of PR8 virus which was 50% of the lethal dose for mice in the 20-day observation period. DAD with alveolar collapse was observed in all dead mice. However, in the surviving mice, instead of DAD, glandular metaplasia was broadly observed in their lungs. The present study indicates that DAD with severe alveolar collapse is associated with death in this mouse infection model of influenza virus. Inhibition of the development of DAD with alveolar collapse may decrease the mortality rate in severe viral pneumonia caused by influenza virus infection.


Subject(s)
Influenza A virus/pathogenicity , Lung/pathology , Lung/virology , Orthomyxoviridae Infections/pathology , Animals , Cell Line , Dogs , Female , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology
12.
Circ Res ; 107(10): 1220-31, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20884878

ABSTRACT

RATIONALE: A homozygous disruption or genetic mutation of the bag3 gene, a member of the Bcl-2-associated athanogene (BAG) family proteins, causes cardiomyopathy and myofibrillar myopathy that is characterized by myofibril and Z-disc disruption. However, the detailed disease mechanism is not yet fully understood. OBJECTIVE: bag3(-/-) mice exhibit differences in the extent of muscle degeneration between muscle groups with muscles experiencing the most usage degenerating at an accelerated rate. Usage-dependent muscle degeneration suggests a role for BAG3 in supporting cytoskeletal connections between the Z-disc and myofibrils under mechanical stress. The mechanism by which myofibrillar structure is maintained under mechanical stress remains unclear. The purpose of the study is to clarify the detailed molecular mechanism of BAG3-mediated muscle maintenance under mechanical stress. METHODS AND RESULTS: To address the question of whether bag3 gene knockdown induces myofibrillar disorganization caused by mechanical stress, in vitro mechanical stretch experiments using rat neonatal cardiomyocytes and a short hairpin RNA-mediated gene knockdown system of the bag3 gene were performed. As expected, mechanical stretch rapidly disrupts myofibril structures in bag3 knockdown cardiomyocytes. BAG3 regulates the structural stability of F-actin through the actin capping protein, CapZß1, by promoting association between Hsc70 and CapZß1. BAG3 facilitates the distribution of CapZß1 to the proper location, and dysfunction of BAG3 induces CapZ ubiquitin-proteasome-mediated degradation. Inhibition of CapZß1 function by overexpressing CapZß2 increased myofibril vulnerability and fragmentation under mechanical stress. On the other hand, overexpression of CapZß1 inhibits myofibrillar disruption in bag3 knockdown cells under mechanical stress. As a result, heart muscle isolated from bag3(-/-) mice exhibited myofibrillar degeneration and lost contractile activity after caffeine contraction. CONCLUSIONS: These results suggest novel roles for BAG3 and Hsc70 in stabilizing myofibril structure and inhibiting myofibrillar degeneration in response to mechanical stress. These proteins are possible targets for further research to identify therapies for myofibrillar myopathy or other degenerative diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CapZ Actin Capping Protein/metabolism , Carrier Proteins/metabolism , HSC70 Heat-Shock Proteins/metabolism , Mechanotransduction, Cellular , Myocardium/metabolism , Myofibrils/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Caffeine/pharmacology , CapZ Actin Capping Protein/genetics , Carrier Proteins/genetics , Cells, Cultured , Humans , Mechanotransduction, Cellular/drug effects , Mice , Mice, Knockout , Myocardial Contraction , Myocardium/pathology , Myofibrils/drug effects , Myofibrils/pathology , Protein Stability , RNA Interference , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Transfection
13.
Biochem Biophys Res Commun ; 401(1): 75-8, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20833141

ABSTRACT

CPI-17 is a unique phosphoprotein that specifically inhibits myosin light chain phosphatase in smooth muscle and plays an essential role in agonist-induced contraction. To elucidate the in situ mechanism for G protein-mediated Ca²+-sensitization of CPI-17 phosphorylation, α-toxin-permeabilized arterial smooth muscle strips were used to monitor both force development and CPI-17 phosphorylation in response to GTPγS with varying Ca²+ concentrations. CPI-17 phosphorylation increased at unphysiologically high Ca²+ levels of pCa ≤ 6. GTPγS markedly enhanced the Ca²+ sensitivity of CPI-17 steady-state phosphorylation but had no enhancing effect under Ca²+-free conditions, while the potent PKC activator PDBu increased CPI-17 phosphorylation regardless of Ca²+ concentration. CPI-17 phosphorylation induced by pCa 4.5 alone was markedly inhibited by the presence of PKC inhibitor but not ROCK inhibitor. In the presence of calyculin A, a potent PP1/PP2A phosphatase inhibitor, CPI-17 phosphorylation increased with time even under Ca²+-free conditions. Furthermore, as Ca²+ concentration increased, so did CPI-17 phosphorylation rate. GTPγS markedly enhanced the rate of phosphorylation of CPI-17 at a given Ca²+. In the absence of calyculin A, either steady-state phosphorylation of CPI-17 under Ca²+-free conditions in the presence of GTPγS or at pCa 6.7 in the absence of GTPγS was negligible, suggesting a high intrinsic CPI-17 phosphatase activity. In conclusion, cooperative increases in Ca²+ and G protein activation are required for a significant activation of total kinases that phosphorylate CPI-17, which together overcome CPI-17 phosphatase activity and effectively increase the Ca²+ sensitivity of CPI-17 phosphorylation and smooth muscle contraction.


Subject(s)
Calcium/metabolism , GTP-Binding Proteins/metabolism , Muscle Contraction , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/physiology , Phosphoproteins/metabolism , Animals , Capillary Permeability , Cells, Cultured , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Muscle, Smooth, Vascular/enzymology , Phosphorylation , Rabbits
14.
J Physiol ; 587(Pt 14): 3587-603, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19470783

ABSTRACT

Nitric oxide (NO) from endothelium is a major mediator of vasodilatation through cGMP/PKG signals that lead to a decrease in Ca(2+) concentration. In addition, NO-mediated signals trigger an increase in myosin light chain phosphatase (MLCP) activity. To evaluate the mechanism of NO-induced relaxation through MLCP deinhibition, we compared time-dependent changes in Ca(2+), myosin light chain (MLC) phosphorylation and contraction to changes in phosphorylation levels of CPI-17 at Thr38, RhoA at Ser188, and MYPT1 at Ser695, Thr696 and Thr853 in response to sodium nitroprusside (SNP)-induced relaxation in denuded rabbit femoral artery. During phenylephrine (PE)-induced contraction, SNP reduced CPI-17 phosphorylation to a minimal value within 15 s, in parallel with decreases in Ca(2+) and MLC phosphorylation, followed by a reduction of contractile force having a latency period of about 15 s. MYPT1 phosphorylation at Ser695, the PKG-target site, increased concurrently with relaxation. Phosphorylation of RhoA, MYPT1 Thr696 and Thr853 differed significantly at 5 min but not within 1 min of SNP exposure. Inhibition of Ca(2+) release delayed SNP-induced relaxation while inhibition of Ca(2+) channel, BK(Ca) channel or phosphodiesterase-5 did not. Pretreatment of resting artery with SNP suppressed an increase in Ca(2+), contractile force and phosphorylation of MLC, CPI-17, MYPT1 Thr696 and Thr853 at 10 s after PE stimulation, but had no effect on phorbol ester-induced CPI-17 phosphorylation. Together, these results suggest that NO production suppresses Ca(2+) release, which causes an inactivation of PKC and rapid CPI-17 dephosphorylation as well as MLCK inactivation, resulting in rapid MLC dephosphorylation and relaxation.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Phosphatase/metabolism , Nitric Oxide/physiology , Phosphoprotein Phosphatases/metabolism , Animals , Calcium/metabolism , Feedback/physiology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Muscle Proteins , Phosphorylation , Rabbits , Vasodilation/physiology
15.
Structure ; 15(12): 1591-602, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18073109

ABSTRACT

Phosphorylation of endogenous inhibitor proteins for type-1 Ser/Thr phosphatase (PP1) provides a mechanism for reciprocal coordination of kinase and phosphatase activities. A myosin phosphatase inhibitor protein CPI-17 is phosphorylated at Thr38 through G-protein-mediated signals, resulting in a >1000-fold increase in inhibitory potency. We show here the solution NMR structure of phospho-T38-CPI-17 with rmsd of 0.36 +/- 0.06 A for the backbone secondary structure, which reveals how phosphorylation triggers a conformational change and exposes an inhibitory surface. This active conformation is stabilized by the formation of a hydrophobic core of intercalated side chains, which is not formed in a phospho-mimetic D38 form of CPI-17. Thus, the profound increase in potency of CPI-17 arises from phosphorylation, conformational change, and hydrophobic stabilization of a rigid structure that poses the phosphorylated residue on the protein surface and restricts its hydrolysis by myosin phosphatase. Our results provide structural insights into transduction of kinase signals by PP1 inhibitor proteins.


Subject(s)
Enzyme Inhibitors/metabolism , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Amino Acid Sequence , Animals , Circular Dichroism , Enzyme Inhibitors/chemistry , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Conformation , Sequence Homology, Amino Acid , Swine
16.
Development ; 134(18): 3327-38, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17720696

ABSTRACT

Heparan sulfate (HS) plays an essential role in extracellular signaling during development. Biochemical studies have established that HS binding to ligands and receptors is regulated by the fine 6-O-sulfated structure of HS; however, mechanisms that control sulfated HS structure and associated signaling functions in vivo are not known. Extracellular HS 6-O-endosulfatases, SULF1 and SULF2, are candidate enzymatic regulators of HS 6-O-sulfated structure and modulate HS-dependent signaling. To investigate Sulf regulation of developmental signaling, we have disrupted Sulf genes in mouse and identified redundant functions of Sulfs in GDNF-dependent neural innervation and enteric glial formation in the esophagus, resulting in esophageal contractile malfunction in Sulf1(-/-);Sulf2(-/-) mice. SULF1 is expressed in GDNF-expressing esophageal muscle and SULF2 in innervating neurons, establishing their direct functions in esophageal innervation. Biochemical and cell signaling studies show that Sulfs are the major regulators of HS 6-O-desulfation, acting to reduce GDNF binding to HS and to enhance GDNF signaling and neurite sprouting in the embryonic esophagus. The functional specificity of Sulfs in GDNF signaling during esophageal innervation was established by showing that the neurite sprouting is selectively dependent on GDNF, but not on neurotrophins or other signaling ligands. These findings provide the first in vivo evidence that Sulfs are essential developmental regulators of cellular HS 6-O-sulfation for matrix transmission and reception of GDNF signal from muscle to innervating neurons.


Subject(s)
Esophagus/embryology , Esophagus/innervation , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Peripheral Nervous System/embryology , Sulfatases/physiology , Sulfotransferases/physiology , Animals , Esophagus/metabolism , Heparitin Sulfate/metabolism , Mice , Mice, Mutant Strains , Muscle Contraction , Muscle, Skeletal/physiology , Myocytes, Smooth Muscle/physiology , Neurites/physiology , Neuroglia/physiology , Neurons/physiology , Signal Transduction , Sulfatases/genetics , Sulfotransferases/genetics
17.
Circ Res ; 100(1): 121-9, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17158339

ABSTRACT

Ca(2+) ion is a universal intracellular messenger that regulates numerous biological functions. In smooth muscle, Ca(2+) with calmodulin activates myosin light chain (MLC) kinase to initiate a rapid MLC phosphorylation and contraction. To test the hypothesis that regulation of MLC phosphatase is involved in the rapid development of MLC phosphorylation and contraction during Ca(2+) transient, we compared Ca(2+) signal, MLC phosphorylation, and 2 modes of inhibition of MLC phosphatase, phosphorylation of CPI-17 Thr38 and MYPT1 Thr853, during alpha(1) agonist-induced contraction with/without various inhibitors in intact rabbit femoral artery. Phenylephrine rapidly induced CPI-17 phosphorylation from a negligible amount to a peak value of 0.38+/-0.04 mol of Pi/mol within 7 seconds following stimulation, similar to the rapid time course of Ca(2+) rise and MLC phosphorylation. This rapid CPI-17 phosphorylation was dramatically inhibited by either blocking Ca(2+) release from the sarcoplasmic reticulum or by pretreatment with protein kinase C inhibitors, suggesting an involvement of Ca(2+)-dependent protein kinase C. This was followed by a slow Ca(2+)-independent and Rho-kinase/protein kinase C-dependent phosphorylation of CPI-17. In contrast, MYPT1 phosphorylation had only a slow component that increased from 0.29+/-0.09 at rest to the peak of 0.68+/-0.14 mol of Pi/mol at 1 minute, similar to the time course of contraction. Thus, there are 2 components of the Ca(2+) sensitization through inhibition of MLC phosphatase. Our results support the hypothesis that the initial rapid Ca(2+) rise induces a rapid inhibition of MLC phosphatase coincident with the Ca(2+)-induced MLC kinase activation to synergistically initiate a rapid MLC phosphorylation and contraction in arteries with abundant CPI-17 content.


Subject(s)
Calcium/metabolism , Femoral Artery/physiology , Muscle, Smooth, Vascular/physiology , Vasoconstriction/physiology , Adrenergic alpha-Agonists/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling , Femoral Artery/drug effects , Femoral Artery/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Phenylephrine/pharmacology , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Subunits/metabolism , Rabbits , Time Factors , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , rho-Associated Kinases
18.
J Cell Sci ; 119(Pt 9): 1769-80, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16608882

ABSTRACT

Phosphorylation of myosin light chain (MLC) and contraction of differentiated smooth muscle cells in vascular walls are regulated by Ca2+ -dependent activation of MLC kinase, and by Rho-kinase- or protein-kinases-C-dependent inhibition of MLC phosphatase (MLCP). We examined regulatory pathways for MLC kinase and MLCP in cultured vascular smooth muscle cells (VSMCs), and for isometric force generation of VSMCs reconstituted in collagen fibers. Protein levels of RhoA, Rho-kinase and MYPT1 (a regulatory subunit of MLCP) were upregulated in cultured VSMCs, whereas a MLCP inhibitor protein, CPI-17, was downregulated. Endothelin-1 evoked a steady rise in levels of Ca2+, MLC phosphorylation and the contractile force of VSMCs, whereas angiotensin-II induced transient signals. Also, Thr853 phosphorylation of MYPT1 occurred in response to stimuli, but neither agonist induced phosphorylation of MYPT1 at Thr696. Unlike fresh aortic tissues, removal of Ca2+ or addition of voltage-dependent Ca2+ -channel blocker did not inhibit contractions of reconstituted VSMC fibers induced by agonists or even high concentrations of extracellular K+ ions. Inhibitors of Ins(1,4,5)P3-receptor and Rho-kinase antagonized agonist-induced or high-K+ -induced contraction in both reconstituted fibers and fresh tissues. These results indicate that both Ins(1,4,5)P3-induced Ca2+ release and Rho-kinase-induced MYPT1 phosphorylation at Thr853 play pivotal roles in MLC phosphorylation of cultured VSMCs where either Ca2+ -influx or CPI-17-MLCP signaling is downregulated.


Subject(s)
Isometric Contraction/physiology , Membrane Potentials/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/metabolism , Signal Transduction/physiology , Angiotensin II/metabolism , Aniline Compounds/metabolism , Animals , Aorta/cytology , Calcium/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Endothelin-1/metabolism , Fluorescent Dyes/metabolism , Male , Myocytes, Smooth Muscle/cytology , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Myosin-Light-Chain Phosphatase/metabolism , Phenylephrine/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinase Inhibitors/metabolism , Protein Phosphatase 1 , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/metabolism , Xanthenes/metabolism
19.
Proc Natl Acad Sci U S A ; 101(24): 8888-93, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15184667

ABSTRACT

Inhibition of myosin phosphatase is critical for agonist-induced contractility of vascular smooth muscle. The protein CPI-17 is a phosphorylation-dependent inhibitor of myosin phosphatase and, in response to agonists, Thr-38 is phosphorylated by protein kinase C, producing a >1,000-fold increase in inhibitory potency. Here, we addressed how CPI-17 could selectively inhibit myosin phosphatase among other protein phosphatase-1 (PP1) holoenzymes. PP1 in cell lysates was separated by sequential affinity chromatography into at least two fractions, one bound specifically to thiophospho-CPI-17, and another bound specifically to inhibitor-2. The MYPT1 regulatory subunit of myosin phosphatase was concentrated only in the fraction bound to thiophospho-CPI-17. This binding was eliminated by addition of excess microcystin-LR to the lysate, showing that binding at the active site of PP1 is required. Phospho-CPI-17 failed to inhibit glycogen-bound PP1 from skeletal muscle, composed primarily of PP1 with the striated muscle glycogen-targeting subunit (G(M)) regulatory subunit. Phospho-CPI-17 was dephosphorylated during assay of glycogen-bound PP1, not MYPT1-associated PP1, even though these two holoenzymes have the same PP1 catalytic subunit. Phosphorylation of CPI-17 in rabbit arteries was enhanced by calyculin A but not okadaic acid or fostriecin, consistent with PP1-mediated dephosphorylation. We propose that CPI-17 binds at the PP1 active site where it is dephosphorylated, but association of MYPT1 with PP1C allosterically retards this hydrolysis, resulting in formation of a complex of MYPT1.PP1C.P-CPI-17, leading to an increase in smooth muscle contraction.


Subject(s)
Enzyme Inhibitors/metabolism , Muscle Proteins/metabolism , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/metabolism , Allosteric Regulation , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Femoral Artery/metabolism , Holoenzymes/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mice , Muscle Proteins/genetics , Muscle Proteins/pharmacology , Muscle, Smooth, Vascular/metabolism , Myosin-Light-Chain Phosphatase/genetics , Myosin-Light-Chain Phosphatase/metabolism , NIH 3T3 Cells , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/genetics , Phosphoproteins/genetics , Phosphoproteins/pharmacology , Phosphorylation , Protein Binding , Protein Phosphatase 1 , Protein Subunits , Proteins/metabolism , Rabbits , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
J Physiol ; 557(Pt 2): 515-28, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15090608

ABSTRACT

Ca(2+) sensitivity of arterial contractility is governed by regulating myosin phosphatase activity in response to agonist stimuli. CPI-17, a myosin phosphatase inhibitor phosphoprotein, is phosphorylated concomitantly with agonist-induced contractile Ca(2+) sensitization in mammalian artery. CPI-17 has not been detected in chicken artery, but is readily detectable in pigeon artery. To evaluate a role of CPI-17, we compared contractility of the arteries of 'CPI-17-deficient' chicken with those of CPI-17-rich rabbit and pigeon, and studied the effect of CPI-17-reconstitution in chicken artery. Other major regulatory/contractile proteins for Ca(2+) sensitization are expressed in both chicken and rabbit arteries. Agonists, such as an alpha(1)-agonist and endothelin-1, produced significant contraction in arteries of all species under physiological Ca(2+)-containing conditions. Depletion of Ca(2+) abolished these contractions in chicken but partially inhibited them in rabbit and pigeon arteries. Unlike CPI-17-rich tissues, chicken arteries exerted little Ca(2+) sensitization in response to alpha(1)-agonist or endothelin-1. GTPgammaS produced a slight Ca(2+) sensitizing effect in chicken artery, but this was significantly smaller compared with CPI-17-rich tissues. A PKC activator (PDBu) did not generate but rather reduced a contraction in both intact and alpha-toxin-permeabilized chicken artery in contrast to a large contraction in CPI-17-rich arteries. Myosin light chain phosphorylation was reduced by PDBu in chicken but elevated in rabbit artery. Addition of recombinant CPI-17 into beta-escin-permeabilized chicken artery restored PDBu-induced and enhanced GTPgammaS-induced Ca(2+) sensitization. Thus, CPI-17 is essential for G protein/PKC-mediated Ca(2+) sensitization in smooth muscle.


Subject(s)
Calcium/metabolism , Chickens/physiology , GTP-Binding Proteins/metabolism , Muscle Proteins/deficiency , Muscle, Smooth/physiology , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Phosphoproteins/deficiency , Protein Kinase C/metabolism , Animals , Aorta , Chickens/metabolism , Columbidae , Male , Muscle Contraction/physiology , Muscle Proteins/biosynthesis , Muscle Proteins/immunology , Muscle, Smooth/enzymology , Myosin Light Chains/metabolism , Phosphoproteins/biosynthesis , Phosphoproteins/immunology , Phosphorylation , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...