Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Nutrients ; 13(12)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34959836

ABSTRACT

Shatavari has long been used as an Ayurvedic herb for women's health, but empirical evidence for its effectiveness has been lacking. Shatavari contains phytoestrogenic compounds that bind to the estradiol receptor. Postmenopausal estradiol deficiency contributes to sarcopenia and osteoporosis. In a randomised double-blind trial, 20 postmenopausal women (68.5 ± 6 years) ingested either placebo (N = 10) or shatavari (N = 10; 1000 mg/d, equivalent to 26,500 mg/d fresh weight shatavari) for 6 weeks. Handgrip and knee extensor strength were measured at baseline and at 6 weeks. Vastus lateralis (VL) biopsy samples were obtained. Data are presented as difference scores (Week 6-baseline, median ± interquartile range). Handgrip (but not knee extensor) strength was improved by shatavari supplementation (shatavari +0.7 ± 1.1 kg, placebo -0.4 ± 1.3 kg; p = 0.04). Myosin regulatory light chain phosphorylation, a known marker of improved myosin contractile function, was increased in VL following shatavari supplementation (immunoblotting; placebo -0.08 ± 0.5 a.u., shatavari +0.3 ± 1 arbitrary units (a.u.); p = 0.03). Shatavari increased the phosphorylation of Aktser473 (Aktser473 (placebo -0.6 ± 0.6 a.u., shatavari +0.2 ± 1.3 a.u.; p = 0.03) in VL. Shatavari supplementation did not alter plasma markers of bone turnover (P1NP, ß-CTX) and stimulation of human osteoblasts with pooled sera (N = 8 per condition) from placebo and shatavari supplementation conditions did not alter cytokine or metabolic markers of osteoblast activity. Shatavari may improve muscle function and contractility via myosin conformational change and further investigation of its utility in conserving and enhancing musculoskeletal function, in larger and more diverse cohorts, is warranted.


Subject(s)
Asparagus Plant , Dietary Supplements , Hand Strength , Phosphorylation/drug effects , Postmenopause/drug effects , Aged , Bone Remodeling/drug effects , Double-Blind Method , Female , Humans , Medicine, Ayurvedic , Middle Aged , Myosin Light Chains/drug effects , Postmenopause/physiology , Quadriceps Muscle/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 317(2): L235-L246, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31116578

ABSTRACT

The cyclic interaction between myosin crossbridges and actin filaments underlies smooth muscle contraction. Phosphorylation of the 20-kDa myosin light chain (MLC20) is a crucial step in activating the crossbridge cycle. Our current understanding of smooth muscle contraction is based on observed correlations among MLC20 phosphorylation, maximal shortening velocity (Vmax), and isometric force over the time course of contraction. However, during contraction there are changes in the extent of phosphorylation of many additional proteins as well as changes in activation of enzymes associated with the signaling pathways. As a consequence, the mechanical manifestation of muscle contraction is likely to change with time. To simplify the study of these relationships, we measured the mechanical properties of airway smooth muscle at different levels of MLC20 phosphorylation at a fixed time during contraction. A simple correlation emerged when time-dependent variables were fixed. MLC20 phosphorylation was found to be directly and linearly correlated with the active stress, stiffness, and power of the muscle; the observed weak dependence of Vmax on MLC20 phosphorylation could be explained by the presence of an internal load in the muscle preparation. These results can be entirely explained by the Huxley crossbridge model. We conclude that when the influence of time-dependent events during contraction is held constant, the basic crossbridge mechanism in smooth muscle is the same as that in striated muscle.


Subject(s)
Muscle Contraction/physiology , Muscle, Skeletal/physiology , Muscle, Smooth, Vascular/physiology , Myosin Light Chains/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/physiology , Animals , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/drug effects , Phosphorylation , Respiratory System/drug effects , Respiratory System/metabolism , Sheep
3.
Arthritis Rheumatol ; 70(11): 1879-1889, 2018 11.
Article in English | MEDLINE | ID: mdl-29781582

ABSTRACT

OBJECTIVE: Immune complex (IC) deposition activates polymorphonuclear neutrophils (PMNs), increases vascular permeability, and leads to organ damage in systemic lupus erythematosus and rheumatoid arthritis. The bioactive lipid sphingosine 1-phosphate (S1P), acting via S1P receptor 1 (S1P1 ), is a key regulator of endothelial cell (EC) barrier function. This study was undertaken to investigate whether augmenting EC integrity via S1P1 signaling attenuates inflammatory injury mediated by ICs. METHODS: In vitro barrier function was assessed in human umbilical vein endothelial cells (HUVECs) by electrical cell-substrate impedance sensing. Phosphorylation of myosin light chain 2 (p-MLC-2) and VE-cadherin staining in HUVECs were assessed by immunofluorescence. A reverse Arthus reaction (RAR) was induced in the skin and lungs of mice with S1P1 deleted from ECs (S1P1 EC-knockout [ECKO] mice) and mice treated with S1P1 agonists and antagonists. RESULTS: S1P1 agonists prevented loss of barrier function in HUVECs treated with IC-activated PMNs. S1P1 ECKO and wild-type (WT) mice treated with S1P1 antagonists had amplified RAR, whereas specific S1P1 agonists attenuated skin and lung RAR in WT mice. ApoM-Fc, a novel S1P chaperone, mitigated EC cell barrier dysfunction induced by activated PMNs in vitro and attenuated lung RAR. Expression levels of p-MLC-2 and disruption of VE-cadherin, each representing manifestations of cell contraction and destabilization of adherens junctions, respectively, that were induced by activated PMNs, were markedly reduced by treatment with S1P1 agonists and ApoM-Fc. CONCLUSION: Our findings indicate that S1P1 signaling in ECs modulates vascular responses to IC deposition. S1P1 agonists and ApoM-Fc enhance the EC barrier, limit leukocyte escape from capillaries, and provide protection against inflammatory injury. The S1P/S1P1 axis is a newly identified target to attenuate tissue responses to IC deposition and mitigate end-organ damage.


Subject(s)
Antigen-Antibody Complex/metabolism , Capillary Permeability/genetics , Endothelial Cells/metabolism , Receptors, Lysosphingolipid/genetics , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Anilides/pharmacology , Animals , Antigens, CD/drug effects , Antigens, CD/metabolism , Apolipoproteins M/pharmacology , Arthus Reaction , Cadherins/drug effects , Cadherins/metabolism , Capillary Permeability/drug effects , Cardiac Myosins/drug effects , Cardiac Myosins/metabolism , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Indans/pharmacology , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Knockout , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Organophosphonates/pharmacology , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Skin/blood supply , Skin/drug effects , Skin/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Thiophenes/pharmacology
4.
J Vet Med Sci ; 80(2): 225-234, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29279465

ABSTRACT

Pectenotoxin-2 (PCTX-2) is one of the polyether macrolide toxins isolated from scallops involved in diarrheic shellfish poisoning via actin depolymerization. In the present study, we examined the bioactive mechanism of PCTX-2 in smooth muscle cells and clarify mode of action of the PCTX-2-induced actin depolymerization using purified skeletal actin. PCTX-2 (300 nM-3 µM) non-selectively inhibited vascular smooth muscle contractions elicited by high K+ or phenylephrine in a dose-dependent manner. However, elevated cytosolic Ca2+ and myosin light chain phosphorylation stimulated by high K+ were only slightly inhibited by PCTX-2. By monitoring the fluorescent intensity of pyrenyl-actin, PCTX-2 was found to inhibit both the velocity and degree of actin polymerization. The critical concentration of G-actin was linearly increased in accordance with the concentration of PCTX-2, indicating sequestration of G-actin with 1 to 1 ratio. The kinetics of F-actin depolymerization by dilution assay indicated that PCTX-2 does not sever F-actin. Transmission electron microscopic and confocal microscopic observations demonstrated that PCTX-2 selectively depolymerized filamentous actin without affecting tublin. In conclusion, PCTX-2 is a potent natural actin depolymerizer which sequesters G-actin without severing F-actin.


Subject(s)
Actins/antagonists & inhibitors , Furans/pharmacology , Pyrans/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Macrolides , Male , Microscopy, Electron, Transmission , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/drug effects , Phenylephrine/pharmacology , Polymerization/drug effects , Rats , Rats, Wistar
5.
J Vet Med Sci ; 80(2): 219-224, 2018 Feb 09.
Article in English | MEDLINE | ID: mdl-29269687

ABSTRACT

Angiotensin II (100 nM) induced bi-phasic increases in cytosolic Ca2+ level ([Ca2+]i) through the activation of angiotensin II type 1 receptor. Pharmacological examinations using 10 µM verapamil, 30 µM La3+, and 1 µM thapsigargin indicated that the first phase of the [Ca2+]i-increase was mediated by Ca2+ release from sarcoplasmic reticulum (SR) and Ca2+ influx independently of voltage dependent Ca2+ channel (VDC). In contrast, the second phase of [Ca2+]i-increase was mediated by Ca2+ influx through VDC. Although both [Ca2+]i and myosin light chain (MLC)-phosphorylation at the first phase was apparently exceeded the threshold for contraction as estimated by high K+-induced responses, there was no appreciable contraction, indicating the dissociation between MLC phosphorylation and force during this phase. In contrast, the second phase of [Ca2+]i was associated with the increases in both MLC phosphorylation and force. These results suggest that angiotensin II is a unique agonist which dissociates MLC-phosphorylation from muscle force during the Ca2+ releases from SR.


Subject(s)
Angiotensin II/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Male , Muscle, Smooth, Vascular/physiology , Myosin Light Chains/metabolism , Rats , Rats, Wistar , Sarcoplasmic Reticulum/drug effects , Thapsigargin/pharmacology , Verapamil/pharmacology
6.
Eur Rev Med Pharmacol Sci ; 21(22): 5239-5246, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29228440

ABSTRACT

OBJECTIVE: To observe the effect of metformin on the tight junction of intestinal epithelial cells and its relevant mechanism. MATERIALS AND METHODS: Caco-2 cell monolayers were incubated with or without tumor necrosis factor-α (TNF-α) (10 ng/mL) in the absence or presence of indicated concentrations of metformin. Transepithelial electrical resistance (TEER) was measured at various time points. Caco-2 cell permeability was assessed using fluorescein permeability test. Immunofluorescence was used to detect the distribution of tight junction protein. Western blotting and Real-Time PCR were used to detect the expression of tight junction protein and Myosin light chain kinase (MLCK)-Myosin light chain (MLC) signaling pathway. RESULTS: Metformin attenuates the effects of TNF-α on Caco-2 cell TEER and paracellular permeability, prevents TNF-α-induced morphological disruption of tight junctions, ameliorates the inhibiting effect of TNF-α on epithelial tight junction-related protein expression and suppresses the TNF-α-induced increase in MLCK production. CONCLUSIONS: Metformin can stabilize and up-regulate tight junction protein by inhibiting MLCK-MLC signaling pathway, thus ameliorating the tight junction of intestinal epithelial cells.


Subject(s)
Epithelial Cells/drug effects , Hypoglycemic Agents/pharmacology , Intestinal Mucosa/drug effects , Metformin/pharmacology , Myosin Light Chains/drug effects , Myosin-Light-Chain Kinase/drug effects , Tight Junctions/drug effects , Caco-2 Cells , Cell Membrane/drug effects , Humans , Intestinal Mucosa/cytology , Signal Transduction/drug effects , Tight Junction Proteins/biosynthesis , Tight Junction Proteins/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
7.
Am J Physiol Endocrinol Metab ; 310(9): E724-33, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26956186

ABSTRACT

Impairment of skeletal muscle function has been associated with changes in ovarian hormones, especially estradiol. To elucidate mechanisms of estradiol on skeletal muscle strength, the hormone's effects on phosphorylation of the myosin regulatory light chain (pRLC) and muscle contractility were investigated, hypothesizing an estradiol-specific beneficial impact. In a skeletal muscle cell line, C2C12, pRLC was increased by 17ß-estradiol (E2) in a concentration-dependent manner. In skeletal muscles of C57BL/6 mice that were E2 deficient via ovariectomy (OVX), pRLC was lower than that from ovary-intact, sham-operated mice (Sham). The reduced pRLC in OVX muscle was reversed by in vivo E2 treatment. Posttetanic potentiation (PTP) of muscle from OVX mice was low compared with that from Sham mice, and this decrement was reversed by acute E2 treatment, demonstrating physiological consequence. Western blot of those muscles revealed that low PTP corresponded with low pRLC and higher PTP with greater pRLC. We aimed to elucidate signaling pathways affecting E2-mediated pRLC using a kinase inhibitor library and C2C12 cells as well as a specific myosin light chain kinase inhibitor in muscles. PI3K/Akt, MAPK, and CamKII were identified as candidate kinases sensitive to E2 in terms of phosphorylating RLC. Applying siRNA strategy in C2C12 cells, pRLC triggered by E2 was found to be mediated by estrogen receptor-ß and the G protein-coupled estrogen receptor. Together, these results provide evidence that E2 modulates myosin pRLC in skeletal muscle and is one mechanism by which this hormone can affect muscle contractility in females.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Myosin Light Chains/drug effects , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Estradiol/metabolism , Estrogen Receptor beta/genetics , Female , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Ovariectomy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , RNA, Small Interfering , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction
8.
J Pharmacol Exp Ther ; 356(1): 53-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26493746

ABSTRACT

Endogenous hydrogen sulfide (H2S) is involved in the regulation of vascular tone. We hypothesized that the lowering of calcium and opening of potassium (K) channels as well as calcium-independent mechanisms are involved in H2S-induced relaxation in rat mesenteric small arteries. Amperometric recordings revealed that free [H2S] after addition to closed tubes of sodium hydrosulfide (NaHS), Na2S, and GYY4137 [P-(4-methoxyphenyl)-P-4-morpholinyl-phosphinodithioic acid] were, respectively, 14%, 17%, and 1% of added amount. The compounds caused equipotent relaxations in isometric myographs, but based on the measured free [H2S], GYY4137 caused more relaxation in relation to released free H2S than NaHS and Na2S in rat mesenteric small arteries. Simultaneous measurements of [H2S] and tension showed that 15 µM of free H2S caused 61% relaxation in superior mesenteric arteries. Simultaneous measurements of smooth muscle calcium and tension revealed that NaHS lowered calcium and caused relaxation of NE-contracted arteries, while high extracellular potassium reduced NaHS relaxation without corresponding calcium changes. In NE-contracted arteries, NaHS (1 mM) lowered the phosphorylation of myosin light chain, while phosphorylation of myosin phosphatase target subunit 1 remained unchanged. Protein kinase A and G, inhibitors of guanylate cyclase, failed to reduce NaHS relaxation, whereas blockers of voltage-gated KV7 channels inhibited NaHS relaxation, and blockers of mitochondrial complex I and III abolished NaHS relaxation. Our findings suggest that low micromolar concentrations of free H2S open K channels followed by lowering of smooth muscle calcium, and by another mechanism involving mitochondrial complex I and III leads to uncoupling of force, and hence vasodilation.


Subject(s)
Calcium/metabolism , Hydrogen Sulfide/pharmacology , Mesenteric Arteries/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Potassium Channels/drug effects , Animals , Electron Transport Complex I/drug effects , Electron Transport Complex III/antagonists & inhibitors , Hydrogen Sulfide/metabolism , In Vitro Techniques , KCNQ Potassium Channels/drug effects , Mesenteric Arteries/metabolism , Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Phosphorylation , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Vasodilation/drug effects
9.
J Pharmacol Toxicol Methods ; 70(2): 152-62, 2014.
Article in English | MEDLINE | ID: mdl-25109759

ABSTRACT

INTRODUCTION: Phosphorylation of myosin light chains is a biochemical readout of smooth muscle cell contraction. α2-Adrenoceptor agonists and antagonists may have important applications in cardiovascular drug development. To assess α2-adrenoceptor-mediated drug effects on vascular smooth muscle contraction, we developed a cell-based assay for the quantitative determination of myosin light chain phosphorylation (pMLC20) in cultured A7r5 smooth muscle cells from rat aorta, transfected to express the human α2B-adrenoceptor (A7r5-α2B cell line). METHODS: In a 96-well format, confluent and serum-starved cells (+/- inhibitor preincubation) were treated with receptor ligands for 5-120 s and the evoked pMLC20 response was monitored with a quantitative in-cell immunoassay, employing time-resolved fluorescence technology. Western blotting, immunofluorescent labelling and intracellular calcium concentration measurements were used for assay validation. RESULTS: The α2-adrenoceptor agonist dexmedetomidine induced rapid, transient and dose-dependent (EC50 30-65 nM) myosin light chain phosphorylation, peaking at 20-45 s with an Emax value of approximately 60% over vehicle control. The endogenous agonist arginine vasopressin produced responses that were comparable to those evoked by dexmedetomidine. Blockers of α2-adrenoceptors, myosin light chain kinase, Gi-proteins, Gßγ subunits, L-type calcium channels and phospholipase C antagonized the dexmedetomidine-evoked myosin light chain phosphorylation, whereas blockers of protein kinase C and protein kinase A potentiated the response to dexmedetomidine. DISCUSSION: The novel method is suitable as a ligand profiling tool to assess the capacity of ligands to evoke or inhibit vascular smooth muscle cell contraction and for investigating the intracellular pathways involved in this process. The assay now allows the quantitative determination of pMLC20 signal induction or inhibition in vascular smooth muscle cells and is superior to conventional Western blotting due to the reduced number of cells required and the potential for measurement of detailed time curves, multiple treatments and replicates on each plate.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/metabolism , Receptors, Adrenergic, alpha-2/analysis , Receptors, Adrenergic, alpha-2/metabolism , Cells, Cultured , Dexmedetomidine/pharmacology , Dose-Response Relationship, Drug , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/drug effects , Phosphorylation/drug effects , Structure-Activity Relationship
10.
Oncol Rep ; 32(4): 1473-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051015

ABSTRACT

4-Amino-2-trifluoromethyl-phenyl retinate (ATPR) is a novel all-trans retinoic acid (ATRA) derivative which was reported to have a superior antitumor effect in breast cancer cells. However, little is known about its antitumor effects on human gastric cancer cells and the mechanisms have not been fully elucidated. The results of the present study suggest that in the human gastric carcinoma cell line BGC-823, ATPR plays a more effective role than ATRA at the same dose in inhibiting proliferation, migration and inducing differentiation after the same treatment time. Furthermore, we investigated the preliminary mechanism of ATPR's anti­migration effect. Immunofluorescence assay demonstrated that claudin-18 positioned from cytoplasm to cell surface following ATPR stimuli. Real-time quantitative RT-PCR and western blot analyses showed that ATPR had significant effects on downregulation of the phosphorylation level of myosin light chain II (MLC II) by suppressing myosin light chain kinase (MLCK) and Rho-associated coiled-coil containing kinase (ROCK), as well as its regulation in the protein expression of RARα and RARß. Moreover, ATPR increased the activity of myosin phosphatase by inhibiting ROCK. Consequently, ATPR showed more promising antitumor effects than ATRA in BGC-823 in vitro, and it may conduct its anti-migration effects by decreasing the phosphorylation level of MLC II, as well as by regulating MLCK and ROCK as downstream target genes.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiac Myosins/drug effects , Cell Movement/drug effects , Myosin Light Chains/drug effects , Retinoids/pharmacology , Stomach Neoplasms/metabolism , Cardiac Myosins/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/drug effects , Myosin-Light-Chain Kinase/metabolism , Phosphorylation/drug effects , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , rho-Associated Kinases/drug effects , rho-Associated Kinases/metabolism
11.
J Ethnopharmacol ; 152(2): 364-71, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24486209

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Schisandra chinensis fruit extract (SCE) has been used as a traditional oriental medicine for treating vascular diseases. However, the pharmacologic effects and mechanisms of SCE on vascular fibrosis are still largely unknown. Transforming growth factor ß1 (TGFß1)-mediated cellular changes are closely associated with the pathogenesis of vascular fibrotic diseases. Particularly, TGFß1 induces actin stress fiber formation that is a crucial mechanism underlying vascular smooth muscle cell (VSMC) migration in response to vascular injury. In this study, we investigated the effect of SCE and its active ingredients on TGFß1-induced stress fiber assembly in A7r5 VSMCs. MATERIALS AND METHODS: To investigate pharmacological actions of SCE and its ingredients on TGFß1-treated VSMCs, we have employed molecular and cell biological technologies, such as confocal microscopy, fluorescence resonance energy transfer, western blotting, and radiometric enzyme analyses. RESULTS: We found that SCE inhibited TGFß1-induced stress fiber formation and cell migration. Schisandrin B (SchB) showed the most prominent effect among the active ingredients of SCE tested. SchB reduced TGFß1-mediated phosphorylation of myosin light chain, and this effect was independent of RhoA/Rho-associated kinase pathway. Fluorescence resonance energy transfer and radiometric enzyme assays confirmed that SchB inhibited myosin light chain kinase activity. We also showed that SchB decreased TGFß1-mediated induction of α-smooth muscle actin by inhibiting Smad signaling. CONCLUSIONS: The present study demonstrates that SCE and its active ingredient SchB suppressed TGFß1-induced stress fiber formation at the molecular level. Therefore, our findings may help future investigations to develop multi-targeted therapeutic strategies that attenuate VSMC migration and vascular fibrosis.


Subject(s)
Lignans/pharmacology , Plant Extracts/pharmacology , Polycyclic Compounds/pharmacology , Schisandra/chemistry , Stress Fibers/drug effects , Actins/drug effects , Actins/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Cyclooctanes/isolation & purification , Cyclooctanes/pharmacology , Fruit , Lignans/isolation & purification , Medicine, East Asian Traditional , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Polycyclic Compounds/isolation & purification , Rats , Signal Transduction/drug effects , Smad Proteins/metabolism , Stress Fibers/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
12.
Vascul Pharmacol ; 59(3-4): 96-102, 2013.
Article in English | MEDLINE | ID: mdl-23916920

ABSTRACT

Numerous studies of acetylcholine (ACh)-induced endothelium-dependent relaxation in arteries have been reported since the original description by Furchgott and Zawadzki (1980). ACh also produces endothelium-independent relaxation. However, it is still unknown whether ACh-induced AMP-activated protein kinase (AMPK) activation can attenuate vasoconstriction in endothelium-denuded rat aorta. Here, we investigated whether ACh may exert a regulatory effect for vascular tone via AMPK activation and its underlying mechanism in vascular smooth muscle cells (VSMCs). Western blotting showed that ACh dose- and time-dependently increased LKB1 and AMPK phosphorylation in VSMCs. The ACh-induced activation of AMPK required muscarinic receptors in VSMCs. LKB1 and AMPK activation by ACh inhibited myosin light-chain kinase (MLCK) and phosphorylated myosin light chain (p-MLC) expression in VSMCs. In addition, a tension study showed the inhibitory effect of ACh-induced AMPK activation on phenylephrine-mediated contraction in endothelium-denuded rat aorta. These data suggest that the ACh-induced activation of AMPK may attenuate vasoconstriction via LKB1-AMPK-dependent mechanism in endothelium-denuded rat aorta.


Subject(s)
AMP-Activated Protein Kinases/drug effects , Acetylcholine/pharmacology , Vasoconstriction/drug effects , Vasodilator Agents/pharmacology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Acetylcholine/administration & dosage , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/drug effects , Myosin-Light-Chain Kinase/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Time Factors , Vasodilator Agents/administration & dosage
13.
Vascul Pharmacol ; 59(5-6): 120-6, 2013.
Article in English | MEDLINE | ID: mdl-23872622

ABSTRACT

Vascular smooth muscle cells (VSMCs) exhibit shrinkage-induced activation of Na(+)/H(+) exchanger isoform 1 (NHE-1) and Na(+), K(+), 2Cl(-) cotransporter (NKCC) under hyperosmotic conditions. To investigate the roles of these ion transporters in vascular smooth muscle force induced by hyperosmotic stress, we tested the effects of 5-(N, N-dimethyl)-amiloride (DMA; NHE inhibitor), cariporide (a selective NHE-1 inhibitor), and bumetanide (NKCC inhibitor) on the contractile response of rat aortic rings to hyperosmolar solutions. NHE inhibitors significantly augmented the maximum force response and contractile sensitivity to hyperosmolar sucrose, NaCl, and glucose in endothelium-denuded rings. Bumetanide elicited a comparatively modest increase in sensitivity. NHE inhibitors blocked the increase in intracellular pH and enhanced the cell volume decrease of cultured VSMCs after exposure to hyperosmolar sucrose. However, DMA had no effect on the increase in cytosolic free Ca(2+) concentration ([Ca(2+)]i) in rat VSMCs and on the increases in phosphorylation of myosin phosphatase target subunit 1 and myosin light chain (MLC) in aortic rings in response to hyperosmolar sucrose. Hyperosmolar sucrose-induced force was significantly attenuated by cytochalasin B in the presence or absence of DMA. Exposure to hyperosmolar sucrose increased the ratio of F- to G-actin; the ratio was further elevated by DMA. These results suggest that the potentiation of hyperosmotic shrinkage by NHE inhibition promotes actin polymerization in VSMCs and augments force production independent of changes in [Ca(2+)]i and MLC phosphorylation.


Subject(s)
Aorta, Thoracic/drug effects , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Vasoconstriction/drug effects , Actins/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Aorta, Thoracic/metabolism , Bumetanide/pharmacology , Calcium/metabolism , Guanidines/pharmacology , Hydrogen-Ion Concentration , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Osmolar Concentration , Phosphorylation/drug effects , Polymerization/drug effects , Rats , Rats, Wistar , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Potassium-Chloride Symporters/drug effects , Sulfones/pharmacology
14.
Eur J Dermatol ; 22(1): 46-53, 2012.
Article in English | MEDLINE | ID: mdl-22370167

ABSTRACT

Although recent clinical reports have indicated that recombinant basic fibroblast growth factor (bFGF) promotes scarless wound healing, the mechanism remains unclear. The present study was carried out to elucidate the mechanisms. The protein levels of cellular α-smooth muscle actin increased at 2-4 days after TGFß treatment alone and at 4 to 6 days after a costimulation of bFGF and TGFß. A spontaneous contraction of stressed myofibroblast-collagen matrix was cancelled by bFGF, which was restored under the presence of C3 exotransferase or Y27632. bFGF stimulation of myofibroblasts as well as fibroblasts elicited a transient Rac and Rho activation. bFGF promoted apoptosis of the myofibroblasts but not of the fibroblasts, even in the presence of two different inhibitors, either LY294002 or an Akt inhibitor. The present study suggests that the phosphatidylinositol-3-kinase to Akt as well as the Rho to Rho kinase signaling pathway is involved in bFGF-promoted myofibroblast apoptosis, and bFGF can promote the scarless wound healing upon the induction of apoptosis of myofibroblasts, but not fibroblasts.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/drug effects , Myofibroblasts/drug effects , Signal Transduction , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Amides/pharmacology , Cells, Cultured , Chromones/pharmacology , Cicatrix/prevention & control , Extracellular Matrix/drug effects , Extracellular Matrix/ultrastructure , Fibrillar Collagens/drug effects , Fibrillar Collagens/ultrastructure , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblasts/metabolism , Humans , Morpholines/pharmacology , Myofibroblasts/metabolism , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pyridines/pharmacology , Wound Healing , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
15.
Peptides ; 32(12): 2436-43, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001227

ABSTRACT

Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase inhibitor, is associated with vascular dysfunction. The polypeptide apelin mediates two major actions on blood vessels. However, their combined effects on vascular function are not fully understood. The present study aimed to determine the effect of apelin-13 on myosin light chain (MLC) phosphorylation in vascular smooth muscle cells (VSMCs) under ADMA-induced endothelial leakage conditions. To assess the increased permeability induced by ADMA, human umbilical vein endothelium cells (HUVECs) were plated in transwell dishes. The FITC-dextran flux and FITC-apelin-13 flux through the endothelial monolayer were measured. To examine the effect of leakage of apelin-13 on MLC phosphorylation in HUVSMCs, transwell dishes were used to establish a coculture system with HUVECs in upper chambers and HUVSMCs in lower chambers. Western blot was performed to assess the phospho-MLC levels. ADMA increased endothelial permeability in a concentration- and time-dependent manner, accompanied by actin stress fiber assembly and intercellular gap formation. When HUVECs were treated with ADMA, the permeability to both macromolecular dextran and micromolecular apelin-13 increased significantly. Both p38 MAPK inhibitor and NADPH oxidase inhibitor could prevent HUVECs from the increased permeability, and the changes of cytoskeleton and intercellular junction, which were induced by ADMA. Apelin-13 passed through the ADMA-stimulated endothelial monolayer and increased the expression of phospho-MLC in VSMCs. These results suggest that ADMA increases endothelial permeability, which may involve the p38 MAPK and NADPH oxidase pathway. Apelin-13 can pass through the damaged endothelial barrier, and acts directly on VSMCs to increase MLC phosphorylation.


Subject(s)
Arginine/analogs & derivatives , Capillary Permeability , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myosin Light Chains/drug effects , Acetophenones/pharmacology , Arginine/adverse effects , Blotting, Western , Cells, Cultured , Coculture Techniques/methods , Dextrans/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Imidazoles/pharmacology , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Muscle, Smooth, Vascular/drug effects , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Phosphorylation , Pyridines/pharmacology , Stress Fibers/drug effects , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Genes Cells ; 14(5): 555-68, 2009 May.
Article in English | MEDLINE | ID: mdl-19371382

ABSTRACT

Phosphorylation of myosin II is thought to play an important role in cytokinesis. Although it is well known that phosphorylated regulatory light chain of myosin II (P-MRLC) localizes along the contractile ring, it is not clear how P-MRLC controls myosin II and F-actin in furrow ingression during cytokinesis. To elucidate roles of P-MRLC in furrow ingression, HeLa cells transfected with EGFP-tagged wild-type or each MRLC mutant were observed using a live-imaging microscope. Time-lapse observation revealed that a delay of furrow ingression was observed in the nonphosphorylatable form of MRLC (AA-MRLC)-expressing cell but not in the wild-type or phospho-mimic MRLC-expressing cell. Among each form of MRLC-expressing cell, the total amount of P-MRLC including phospho-mimic MRLCs was smallest in the cell expressing AA-MRLC. However, the amount of F-actin and myosin II at the contractile ring in the AA-MRLC-expressing cell was the same as that in the normal cell. Interestingly, delay of furrow ingression by a Rho-kinase inhibitor, Y27632, was rescued by phospho-mimic MRLCs. These results suggest that the P-MRLC is essential for the progress of furrow ingression but not the retainment of F-actin and myosin II in the contractile ring of dividing HeLa cells.


Subject(s)
Cytokinesis/physiology , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Actins/drug effects , Actins/metabolism , Amides/pharmacology , Cytokinesis/drug effects , HeLa Cells , Humans , Myosin Light Chains/drug effects , Myosin Type II/drug effects , Phosphorylation , Pyridines/pharmacology
17.
Surg Neurol ; 72(5): 490-4; discussion 494-5, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19147193

ABSTRACT

BACKGROUND: The aim of this study was to investigate the ability of a SERM, RLX, to prevent vasospasm in a rabbit model of SAH. METHODS: Thirty-four New Zealand white rabbits were allocated into 3 groups randomly. Subarachnoid hemorrhage was induced by injecting autologous blood into the cisterna magna. The treatment groups were as follows: (1) sham operated (no SAH [n = 12]), (2) SAH only (n = 12), and (3) SAH plus RLX (n = 10). Basilar artery lumen areas and arterial wall thickness were measured to assess vasospams in all groups. RESULTS: There was a statistically significant difference between the mean basilar artery cross-sectional areas and the mean arterial wall thickness measurements of the control and SAH-only groups (P < .05). The difference between the mean basilar artery cross-sectional areas and the mean arterial wall thickness measurements in the RLX-treated group was statistically significant (P < .05). The difference between the SAH group and the SAH + RLX group was also statistically significant (P < .05). CONCLUSIONS: These findings demonstrate that RLX has marked vasodilatatory effect in an experimental model of SAH in rabbits. This observation may have clinical implications suggesting that this SERM drug could be used as possible anti-vasospastic agent in patients without major adverse effects.


Subject(s)
Cerebral Arteries/drug effects , Raloxifene Hydrochloride/pharmacology , Subarachnoid Hemorrhage/complications , Vasodilator Agents/pharmacology , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/etiology , Animals , Basilar Artery/drug effects , Basilar Artery/pathology , Basilar Artery/physiopathology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cyclic GMP/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/metabolism , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Rabbits , Raloxifene Hydrochloride/therapeutic use , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use , Treatment Outcome , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/therapeutic use , Vasospasm, Intracranial/physiopathology
18.
Eur J Pharmacol ; 599(1-3): 137-45, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-18929558

ABSTRACT

Urinary bladder wall muscle (i.e., detrusor smooth muscle; DSM) contracts in response to a quick-stretch, but this response is neither fully characterized, nor completely understood at the subcellular level. Strips of rabbit DSM were quick-stretched (5 ms) and held isometric for 10 s to measure the resulting peak quick-stretch contractile response (PQSR). The ability of selective Ca(2+) channel blockers and kinase inhibitors to alter the PQSR was measured, and the phosphorylation levels of myosin light chain (MLC) and myosin phosphatase targeting regulatory subunit (MYPT1) were recorded. DSM responded to a quick-stretch with a biphasic response consisting of an initial contraction peaking at 0.24+/-0.02-fold the maximum KCl-induced contraction (F(o)) by 1.48+/-0.17 s (PQSR) before falling to a weaker tonic (10 s) level (0.12+/-0.03-fold F(o)). The PQSR was dependent on the rate and degree of muscle stretch, displayed a refractory period, and was converted to a sustained response in the presence of muscarinic receptor stimulation. The PQSR was inhibited by nifedipine, 2-aminoethoxydiphenyl borate (2-APB), 100 microM gadolinium and Y-27632, but not by atropine, 10 microM gadolinium, LOE-908, cyclopiazonic acid, or GF-109203X. Y-27632 and nifedipine abolished the increase in MLC phosphorylation induced by a quick-stretch. Y-27632, but not nifedipine, inhibited basal MYPT1 phosphorylation, and a quick-stretch failed to increase phosphorylation of this rhoA kinase (ROCK) substrate above the basal level. These data support the hypothesis that constitutive ROCK activity is required for a quick-stretch to activate Ca(2+) entry and cause a myogenic contraction of DSM.


Subject(s)
Calcium/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , rho-Associated Kinases/metabolism , Animals , Calcium Channel Blockers/pharmacology , Female , In Vitro Techniques , Isometric Contraction/physiology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Potassium Chloride/pharmacology , Protein Kinase Inhibitors/pharmacology , Rabbits , Urinary Bladder/drug effects , Urinary Bladder/metabolism , rho-Associated Kinases/drug effects
19.
J Cell Physiol ; 217(1): 72-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18498123

ABSTRACT

Treating rats with vanadate, a nonspecific inhibitor of protein tyrosine phosphatases, optimizes the uniform packing of collagen fiber bundles in wound granulation tissue and doubles wound breaking strength in rat incisional wounds. The speculation is vanadate optimizes the packing of collagen fiber bundles through the orientation of newly arrived wound fibroblasts in the fibrin clot filling the defect. Segments of 14 day chick embryo tendons were placed on fibrin clots and maintained in organ culture with and without 30 microM vanadate. On day 7 explants were examined histologically and biochemically. Tendon fibroblast outgrowth from untreated explants migrated in a random fashion, while fibroblasts from vanadate-treated explants migrated out in linear arrays. Fibroblasts were elongated by 20% form vanadate treated explant compared to controls. Myosin ATPase, required for optimal cell motility, is optimized by the phosphorylation of its myosin light chain (MLC). Western blot analysis of lysates from the fibroblasts that migrated into the fibrin showed vanadate increased MLC-P levles. These findings support the notion that vanadate promotes the deposition of regular, parallel collagen fiber bundles by advancing the orientation of fibroblasts in parallel linear arrays early in the wound repair process.


Subject(s)
Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Fibrin/metabolism , Fibroblasts/drug effects , Vanadates/pharmacology , Wound Healing/drug effects , Animals , Blotting, Western , Chick Embryo , Extracellular Matrix/chemistry , Fibroblasts/cytology , Humans , Microscopy, Fluorescence , Myosin Light Chains/drug effects , Organ Culture Techniques , Phosphorylation , Tendons/cytology , Wound Healing/physiology
20.
Exp Toxicol Pathol ; 60(1): 9-15, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18434112

ABSTRACT

Airway hyperresponsiveness (AHR) is a hallmark of bronchial asthma. Increased expression of smooth muscle contractile proteins or increased responsiveness of the contractile apparatus due to RhoA/Rho-kinase activation may contribute to AHR. BALB/c mice developed AHR following systemic sensitization by intraperitoneal injections of 20 microg ovalbumin (OVA) in presence of 2mg Al(OH)(3) on days 1 and 14, and airway challenge by 1% OVA-inhalation for 20 min each on days 28, 29 and 30. As assessed by Western blot, protein expression of RhoA, MLC (myosin light chain) and smMLCK (smooth muscle myosin light chain kinase) was increased in lungs of OVA/OVA-animals with AHR, as well as in lungs of OVA-sensitized and sham-challenged animals (OVA/PBS) without AHR, compared with lungs of PBS/PBS-animals. Pretreatment with the specific Rho-kinase inhibitor Y-27632 reduced MLC-phosphorylation and AHR. Contribution of Rho-kinase to bronchoconstriction was increased in lungs of OVA/OVA-animals compared with OVA/PBS- and PBS/PBS-animals, respectively. Furthermore, bronchoconstriction following MCh stimulation was significantly reduced after Y-27632 application. In conclusion, systemic allergen-sensitization increased pulmonary expression of proteins involved in smooth muscle contraction, which may contribute to development of AHR. However, this observation was independent from local allergen challenge, suggesting that additional cofactors may be required for the activation of Rho-kinase and thereby the induction of AHR. Rho-kinase may play an important role in murine AHR, and the bronchodilating action of Rho-kinase inhibition may offer a new therapeutic perspective in obstructive airway disease.


Subject(s)
Asthma/enzymology , Bronchial Hyperreactivity/enzymology , Bronchoconstriction/physiology , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , rho-Associated Kinases/metabolism , Amides/pharmacology , Animals , Asthma/chemically induced , Asthma/drug therapy , Blotting, Western , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/drug therapy , Bronchoconstriction/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Lung/drug effects , Lung/enzymology , Mice , Mice, Inbred BALB C , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Myosin Light Chains/drug effects , Myosin-Light-Chain Kinase/drug effects , Ovalbumin/immunology , Perfusion , Phosphorylation , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...