Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Publication year range
2.
Basic Res Cardiol ; 110(4): 38, 2015.
Article in English | MEDLINE | ID: mdl-25982880

ABSTRACT

Recent studies demonstrated that NADPH oxidase 2 (NOX2) expression in myocardium after ischemia-reperfusion (IR) is significantly upregulated. However, the underlying mechanisms remain unknown. This study aims to determine if nuclear cardiac myosin light chain 2 (MYL2), a well-known regulatory subunit of myosin, functions as a transcription factor to promote NOX2 expression following myocardial IR in a phosphorylation-dependent manner. We examined the phosphorylation status of nuclear MYL2 (p-MYL2) in a rat model of myocardial IR (left main coronary artery subjected to 1 h ligation and 3 h reperfusion) injury, which showed IR injury and upregulated NOX2 expression as expected, accompanied by elevated H2O2 and nuclear p-MYL2 levels; these effects were attenuated by inhibition of myosin light chain kinase (MLCK). Next, we explored the functional relationship of nuclear p-MYL2 with NOX2 expression in H9c2 cell model of hypoxia-reoxygenation (HR) injury. In agreement with our in vivo findings, HR treatment increased apoptosis, NOX2 expression, nuclear p-MYL2 and H2O2 levels, and the increases were ameliorated by inhibition of MLCK or knockdown of MYL2. Finally, molecular biology techniques including co-immunoprecipitation (Co-IP), chromatin immunoprecipitation (ChIP), DNA pull-down and luciferase reporter gene assay were utilized to decipher the molecular mechanisms. We found that nuclear p-MYL2 binds to the consensus sequence AGCTCC in NOX2 gene promoter, interacts with RNA polymerase II and transcription factor IIB to form a transcription preinitiation complex, and thus activates NOX2 gene transcription. Our results demonstrate that nuclear MYL2 plays an important role in IR injury by transcriptionally upregulating NOX2 expression to enhance oxidative stress in a phosphorylation-dependent manner.


Subject(s)
Cardiac Myosins/physiology , Membrane Glycoproteins/genetics , Myocardium/metabolism , Myosin Light Chains/physiology , NADPH Oxidases/genetics , Animals , Cardiac Myosins/analysis , Cell Nucleus/chemistry , Cells, Cultured , Male , Myocardial Reperfusion Injury/prevention & control , Myosin Light Chains/analysis , Myosin-Light-Chain Kinase/antagonists & inhibitors , NADPH Oxidase 2 , Oxidative Stress , Phosphorylation , Rats , Rats, Sprague-Dawley
3.
Biotechnol Lett ; 36(12): 2581-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25216643

ABSTRACT

To investigate the effect of BMP4 on cardiomyocyte differentiation of adipose tissue-derived stem cells (ADSCs), mouse ADSCs were treated with different concentrations of BMP4 in media containing fetal bovine serum (FBS) or Knockout™ Serum Replacement (KoSR). 3 weeks after cardiac induction, differentiated ADSCs expressed some cardiac-specific genes and proteins. BMP4 treatment upregulated the expression of cardiac transcription factors. In both FBS and KoSR-supplemented media, lower concentrations of BMP4 had a positive effect on the expression of MLC2A gene, while MLC2V was more expressed with higher concentrations of BMP4. BMP4 treatment in KoSR supplemented medium was more efficient for cardiac induction. Supplementation of culture media with insulin-transferrin-selenium improved the expression of MLC2A gene. The results of this study indicated that BMP4 is important for cardiac differentiation of the ADSCs. However, BMP4 was not enough for structural and functional maturation of the ADSC-derived cardiomyocytes.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/drug effects , Stem Cells/drug effects , Stem Cells/physiology , Animals , Cells, Cultured , Culture Media/chemistry , Gene Expression Profiling , Humans , Mice , Myosin Light Chains/analysis , Myosin Light Chains/genetics
4.
Int J Sports Med ; 35(6): 476-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24234010

ABSTRACT

Evidence suggests that myofibers from endurance trained skeletal muscle display unique contractile parameters. However, the underlying mechanisms remain unclear. To further elucidate the influence of endurance training on myofiber contractile function, we examined factors that may impact myofilament interactions (i. e., water content, concentration of specific protein fractions, actin and myosin content) or directly modulate myosin heavy chain (MHC) function (i. e., myosin light chain (MLC) composition) in muscle biopsy samples from highly-trained competitive (RUN) and recreational (REC) runners. Muscle water content was lower (P<0.05) in RUN (73±1%) compared to REC (75±1%) and total muscle and myofibrillar protein concentration was higher (P<0.05) in RUN, which may indicate differences in myofilament spacing. Content of the primary contractile proteins, myosin (0.99±0.08 and 1.01±0.07 AU) and actin (1.33±0.09 and 1.27±0.09 AU) in addition to the myosin to actin ratio (0.75±0.04 and 0.80±0.06 AU) was not different between REC and RUN, respectively, when expressed relative to the amount of myofibrillar protein. At the single-fiber level, slow-twitch MHC I myofibers from RUN contained less (P<0.05) MLC 1 and greater (P<0.05) amounts of MLC 3 than REC, while MLC composition was similar in fast-twitch MHC IIa myofibers between REC and RUN. These data suggest that the distinctive myofiber contractile profile in highly-trained runners may be partially explained by differences in the content of the primary contractile proteins and provides unique insight into the modulation of contractile function with extreme loading -patterns.


Subject(s)
Actins/analysis , Myofibrils/chemistry , Myosin Heavy Chains/analysis , Myosin Light Chains/analysis , Physical Endurance/physiology , Running/physiology , Actins/metabolism , Adult , Biopsy , Body Water/metabolism , Humans , Muscle Contraction , Myofibrils/metabolism , Myosin Heavy Chains/metabolism , Myosin Light Chains/metabolism , Young Adult
5.
Biochemistry ; 52(9): 1622-30, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23339370

ABSTRACT

We have used time-resolved phosphorescence anisotropy to investigate the effects of essential light chain (ELC) isoforms (A1 and A2) on the interaction of skeletal muscle myosin with actin, to relate structural dynamics to previously reported functional effects. Actin was labeled with a phosphorescent probe at C374, and the myosin head (S1) was separated into isoenzymes S1A1 and S1A2 by ion-exchange chromatography. As previously reported, S1A1 exhibited substantially lower ATPase activity at saturating actin concentrations but substantially higher apparent actin affinity, resulting in a higher catalytic efficiency. In the absence of ATP, each isoenzyme increased actin's final anisotropy cooperatively and to a similar extent, indicating a similar restriction of the amplitude of intrafilament rotational motions in the strong-binding (S) state of actomyosin. In contrast, in the presence of a saturating level of ATP, S1A1 increased actin anisotropy much more than S1A2 and with greater cooperativity, indicating that S1A1 was more effective in restricting actin dynamics during the active interaction of actin and myosin. We conclude that during the active interaction of actin and ATP with myosin, S1A1 is more effective at stabilizing the S state (probably the force-generating state) of actomyosin, while S1A2 tends to stabilize the weak-binding (non-force-generating) W state. When a mixture of isoenzymes is present, S1A1 is dominant in its effects on actin dynamics. We conclude that ELC of skeletal muscle myosin modulates strong-to-weak structural transitions during the actomyosin ATPase cycle in an isoform-dependent manner, with significant implications for the contractile function of actomyosin.


Subject(s)
Actins/metabolism , Myosin Light Chains/metabolism , Actins/analysis , Actomyosin/analysis , Actomyosin/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Anisotropy , Luminescent Agents/analysis , Luminescent Measurements , Models, Molecular , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Myosin Light Chains/analysis , Osmolar Concentration , Protein Isoforms/analysis , Protein Isoforms/metabolism , Rabbits
6.
Proteomics ; 12(18): 2879-89, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22851476

ABSTRACT

Investigation of protein changes as well as authentication of meat is particularly difficult in processed meat products due to their different composition, complexity and very often inhomogeneity. The aim of this study was to check if the inter-species differences in the expression of myosin light chain (MLC) isoforms observed in raw meat were retained in meat products. MLCs from mixtures of minced meat (16 variants), frankfurters and sausages (15 products) made from cattle, pig, chicken, turkey, duck and goose were analysed by 2DE. Species-specific patterns of MLC isoforms were observed in all the mixtures and processed meat products. Relatively small degradation was observed in the MLCs after processing. Image analysis enabled species identification of the meat in all samples when the content of meat of one species was not lower than 10%. However, it was impossible to differentiate between all the six species under investigation on the basis of individual isoform. It was possible when the combination of all the three isoforms (myosin light chain 1 fast, myosin light chain 2 fast and myosin light chain 3 fast) was analysed. The results evidenced that MLCs have potential to be used as markers in authentication of meat products made from the analysed six species.


Subject(s)
Electrophoresis, Gel, Two-Dimensional , Meat/analysis , Myosin Light Chains/analysis , Amino Acid Sequence , Animals , Cattle , Chickens , Ducks , Meat Products/analysis , Molecular Sequence Data , Protein Isoforms/analysis , Sus scrofa , Turkeys
7.
J Proteome Res ; 9(7): 3374-83, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20433202

ABSTRACT

A proteomic-based method has been developed for the detection of chicken meat within mixed meat preparations. The procedure is robust and simple, comprising the extraction of myofibrillar proteins, enrichment of target proteins using OFFGEL isoelectric focusing, in-solution trypsin digestion of myosin light chain 3, and analysis of the generated peptides by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). Using this approach, it was possible for example to detect 0.5% contaminating chicken in pork meat with high confidence. Quantitative detection of chicken meat was done by using AQUA stable isotope peptides made from the sequence of previously selected species-specific peptide biomarkers. Linearity was observed between the amount of the peptide biomarker and the amount of chicken present in the mixture; further independent replication is required now to validate the method. Apart from its simplicity, this approach has the advantage that it can be used effectively for the detection of both raw and cooked meat. The method is robust, reliable, and sensitive, representing a serious alternative to methods currently in use for these purposes. It is amenable to highly processed foods which can be particularly problematic, as the tertiary protein structure is often affected in processed food precluding immunoassays. In addition, this proteomic analysis will permit the determination of definitive discriminatory sequence, unlike the DNA PCR based methods used presently. The present article also demonstrates the translation of the technology to routine mass spectrometry equipment, making the methodology suitable for public analysts.


Subject(s)
Chickens , Food Analysis/methods , Food Contamination/analysis , Meat Products/analysis , Proteomics/methods , Animals , Biomarkers/analysis , Biomarkers/chemistry , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Isotope Labeling , Myosin Light Chains/analysis , Myosin Light Chains/chemistry , Myosin Light Chains/classification , Peptide Fragments/analysis , Peptide Fragments/chemistry , Peptide Fragments/classification , Species Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Trypsin
8.
Mol Biol Cell ; 18(2): 605-16, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17151359

ABSTRACT

Nonmuscle myosin II, an actin-based motor protein, plays an essential role in actin cytoskeleton organization and cellular motility. Although phosphorylation of its regulatory light chain (MRLC) is known to be involved in myosin II filament assembly and motor activity in vitro, it remains unclear exactly how MRLC phosphorylation regulates myosin II dynamics in vivo. We established clones of Madin Darby canine kidney II epithelial cells expressing MRLC-enhanced green fluorescent protein or its mutants. Time-lapse imaging revealed that both phosphorylation and dephosphorylation are required for proper dynamics of myosin II. Inhibitors affecting myosin phosphorylation and MRLC mutants indicated that monophosphorylation of MRLC is required and sufficient for maintenance of stress fibers. Diphosphorylated MRLC stabilized myosin II filaments and was distributed locally in regions of stress fibers where contraction occurs, suggesting that diphosphorylation is involved in the spatial regulation of myosin II assembly and contraction. We further found that myosin phosphatase or Zipper-interacting protein kinase localizes to stress fibers depending on the activity of myosin II ATPase.


Subject(s)
Myosin Light Chains/metabolism , Myosin Type II/metabolism , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/metabolism , Animals , Apoptosis Regulatory Proteins , Calcium-Calmodulin-Dependent Protein Kinases , Cells, Cultured , Death-Associated Protein Kinases , Dogs , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Mutation , Myosin Light Chains/analysis , Myosin Light Chains/genetics , Myosin Type II/analysis , Myosin Type II/genetics , Myosin-Light-Chain Phosphatase/analysis , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism , Stress Fibers/enzymology , Stress Fibers/metabolism , Stress Fibers/ultrastructure
9.
Oxid Med Cell Longev ; 2020: 6427284, 2020.
Article in English | MEDLINE | ID: mdl-33123314

ABSTRACT

An essential procedure for the treatment of myocardial infarction is restoration of blood flow in the obstructed infarct artery, which may cause ischaemia/reperfusion (I/R) injury. Heart I/R injury manifests in oxidative stress, metabolic and morphological disorders, or cardiac contractile dysfunction. Klotho protein was found to be produced in the heart tissue and participate in antioxidation or ion homeostasis. The aim of this study was to examine an influence of Klotho protein on the heart subjected to I/R injury. Wistar rats served as a surrogate heart model ex vivo. Rat hearts perfused using the Langendorff method were subjected to global no-flow ischaemia, and isolated rat cardiomyocytes underwent chemical I/R in vitro, with or without recombinant Klotho protein administration. Haemodynamic parameters of heart function, cell contractility, markers of I/R injury and oxidative stress, and the level of contractile proteins such as myosin light chain 1 (MLC1) and troponin I (TnI) were measured. The treatment of hearts subjected to I/R injury with Klotho protein resulted in a recovery of heart mechanical function and ameliorated myocyte contractility. This improvement was associated with decreased tissue injury, enhanced antioxidant capacity, and reduced release of MLC1 and TnI. The present research showed the contribution of Klotho to cardioprevention during I/R. Thus, Klotho protein may support the protection from I/R injury and prevention of contractile dysfunction in the rat heart.


Subject(s)
Glucuronidase/pharmacology , Heart/drug effects , Myocardium/metabolism , Animals , Antioxidants/chemistry , Glucuronidase/genetics , Glucuronidase/metabolism , Heart/physiology , Heart Rate/drug effects , In Vitro Techniques , Klotho Proteins , Male , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myosin Light Chains/analysis , Oxidative Stress/drug effects , Rats , Rats, Wistar , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Troponin I/analysis
10.
Biochemistry ; 48(4): 754-65, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19127992

ABSTRACT

Myosin 2 is the molecular motor in muscle. It binds actin and executes a power stroke by rotating its lever arm through an angle of approximately 70 degrees to translate actin against resistive force. Myosin 2 has evolved to function optimally under crowded conditions where rates and equilibria of macromolecular reactions undergo major shifts relative to those measured in dilute solution. Hence, an important research objective is to detect in situ the lever arm orientation. Single-molecule measurements are preferred because they clarify ambiguities that are unavoidable with ensemble measurements; however, detecting single molecules in the condensed tissue medium where the myosin concentration exceeds 100 muM is challenging. A myosin light chain (MLC) tagged with photoactivatable green fluorescent protein (PAGFP) was constructed. The recombinant MLC physically and functionally replaced native MLC on the myosin lever arm in a permeabilized skeletal muscle fiber. Probe illumination volume was minimized using total internal reflection fluorescence microscopy, and PAGFP was sparsely photoactivated such that polarized fluorescence identified a single probe orientation. Several physiological states of the muscle fiber were characterized, revealing two distinct orientation populations in all states called straight and bent conformations. Conformation occupancy probability varies among fiber states with rigor and isometric contraction at extremes where straight and bent conformations predominate, respectively. Comparison to previous work on single rigor cross-bridges at the A-band periphery where the myosin concentration is low suggests molecular crowding in the A-band promotes occupancy of the straight myosin conformation [Burghardt, T. P., et al. (2007) Biophys. J. 93, 2226]. The latter may have a role in contraction because it provides additional free energy favoring completion of the cross-bridge power stroke.


Subject(s)
Green Fluorescent Proteins/metabolism , Muscle Fibers, Skeletal/chemistry , Myosin Light Chains/chemistry , Amino Acid Substitution/genetics , Animals , Catalytic Domain/genetics , Crystallography, X-Ray , Fluorescence Polarization , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Myocardium/chemistry , Myocardium/metabolism , Myosin Light Chains/analysis , Myosin Light Chains/genetics , Photochemistry/methods , Rabbits , Swine
11.
Anal Chem ; 81(10): 3950-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19388669

ABSTRACT

Low-abundance protein quantification has historically been performed using ligand binding techniques. However, due to the time and cost associated with developing enzyme-linked immunosorbent assay (ELISA), mass spectrometric approaches are playing an increasingly important role. Protein quantification at or below the nanogram per milliliter level using liquid chromatography/tandem mass spectrometry (LC/MS/MS) typically utilizes an immunoaffinity (IA) enrichment step such as immunoprecipitation. In order to maximize mass spectrometry (MS) sensitivity, protein enrichment is followed by a proteolytic cleavage step used to generate a surrogate peptide with better mass spectrometric properties. Unlike ELISA, IA-LC/MS/MS is a serial technique that can require up to 3 days for a single batch analysis due to lengthy incubation and digestion steps. This report describes the use of immunoprecipitation in 96-well ELISA format (IPE) and microwave-assisted protein digestion to reduce the time required to perform LC/MS/MS protein analyses to within a single day. The utility of this approach was investigated through its application to previously published LC/MS/MS protein assays from our laboratory for two cardiotoxicity biomarkers, Myl3 and NTproBNP. Using commercially available antibodies, IPE and microwave-assisted digestion were used to repeat intraday validations for these markers, and intraday precision (%CV) and accuracy (%RE) did not exceed 11% or 3% for either assay, respectively. Additionally, lower limits of quantification of 100 pg/mL (NTproBNP) and 0.95 ng/mL (Myl3) were achieved.


Subject(s)
Chromatography, Liquid/methods , Myosin Light Chains/analysis , Natriuretic Peptide, Brain/analysis , Peptide Fragments/analysis , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/immunology , Biomarkers/analysis , Humans , Immunoprecipitation , Microwaves , Rats
12.
J Cell Biol ; 140(1): 119-29, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9425160

ABSTRACT

Phosphorylation of the regulatory light chain of myosin II (RMLC) at Serine 19 by a specific enzyme, MLC kinase, is believed to control the contractility of actomyosin in smooth muscle and vertebrate nonmuscle cells. To examine how such phosphorylation is regulated in space and time within cells during coordinated cell movements, including cell locomotion and cell division, we generated a phosphorylation-specific antibody. Motile fibroblasts with a polarized cell shape exhibit a bimodal distribution of phosphorylated myosin along the direction of cell movement. The level of myosin phosphorylation is high in an anterior region near membrane ruffles, as well as in a posterior region containing the nucleus, suggesting that the contractility of both ends is involved in cell locomotion. Phosphorylated myosin is also concentrated in cortical microfilament bundles, indicating that cortical filaments are under tension. The enrichment of phosphorylated myosin in the moving edge is shared with an epithelial cell sheet; peripheral microfilament bundles at the leading edge contain a higher level of phosphorylated myosin. On the other hand, the phosphorylation level of circumferential microfilament bundles in cell-cell contacts is low. These observations suggest that peripheral microfilaments at the edge are involved in force production to drive the cell margin forward while microfilaments in cell-cell contacts play a structural role. During cell division, both fibroblastic and epithelial cells exhibit an increased level of myosin phosphorylation upon cytokinesis, which is consistent with our previous biochemical study (Yamakita, Y., S. Yamashiro, and F. Matsumura. 1994. J. Cell Biol. 124:129-137). In the case of the NRK epithelial cells, phosphorylated myosin first appears in the midzones of the separating chromosomes during late anaphase, but apparently before the formation of cleavage furrows, suggesting that phosphorylation of RMLC is an initial signal for cytokinesis.


Subject(s)
Cell Cycle/physiology , Myosins/metabolism , Phosphoserine , Amino Acid Sequence , Animals , Antibodies , Cell Division , Cell Line , Cell Movement/physiology , Epithelial Cells , Fibroblasts , Interphase , Kidney , Mitosis/physiology , Myosin Light Chains/analysis , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Myosins/analysis , Organelles/physiology , Organelles/ultrastructure , Phosphopeptides/chemistry , Phosphopeptides/immunology , Rats
13.
Cardiovasc Res ; 77(1): 211-20, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18006454

ABSTRACT

AIMS: High-risk atheromatous plaques contain significant extra- and intracellular lipid deposits and very low smooth muscle cell numbers in the intima. However, the mechanisms inducing vessel wall remodelling and high-risk plaque composition are unknown. Low-density lipoproteins (LDLs) infiltrate the vessel wall and become retained and aggregated (agLDL) in the intima by binding to extracellular matrix proteoglycans. The cellular responses triggered by agLDL are not fully understood. This study was designed to investigate the effects of agLDL on vascular remodelling and repair, specifically studying human coronary vascular smooth muscle cell (VSMC) functions. METHODS AND RESULTS: Using a wound repair VSMC model system, we have shown that agLDL significantly impair cell migration. Proteomic analysis revealed a differential phenotypic pattern of myosin light chain and lower levels of phosphorylated myosin regulatory light chain (P-MRLC) in agLDL-exposed VSMC. LDL also induced changes in the subcellular localization of P-MRLC, with dephosphorylation strongly evident on the front edge of agLDL-treated migrating cells. PMA, a strong inducer of myosin light chain (MLC) phosphorylation, significantly reduced the effects of agLDL in VSMC migration. Inhibition of MLC kinase with ML9 did not affect MRLC dephosphorylation already induced by agLDL. CONCLUSION: Our results indicate that LDLs impair human VSMC migration and wound repair after injury. agLDL, and to a lesser extent nLDL, induce dephosphorylation of MRLC and striking changes in the subcellular localization of P-MRLC, a cytoskeleton protein involved in VSMC migration kinetics.


Subject(s)
Coronary Vessels/drug effects , Lipoproteins, LDL/toxicity , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myosin Light Chains/analysis , Proteomics , Cell Movement/drug effects , Cells, Cultured , Coronary Vessels/chemistry , Coronary Vessels/cytology , Cytoskeletal Proteins/analysis , Humans , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/physiology , Myosin-Light-Chain Kinase/physiology , Myosin-Light-Chain Phosphatase/physiology , Phosphorylation , Wound Healing
14.
Biochem Biophys Res Commun ; 374(2): 356-60, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18638453

ABSTRACT

Here, we describe the first example of a cell-based myosin light chain phosphorylation assay in 96-well format that allows for the rapid screening of novel Rho-kinase inhibitors. We obtained IC(50) values for the prototypic Rho-kinase inhibitors Y-27632 (1.2+/-0.05microM) and Fasudil (3.7+/-1.2microM) that were similar to those previously published utilizing electrophoresis-based methodologies. H-1152P, a Fasudil analog showed an IC(50) value of 77+/-30nM. Data derived from a set of 21 novel Rho-kinase inhibitors correlate with those generated by a well-established cell-based phenotypic Rho-kinase inhibition assay (R(2)=0.744). These results show that imaging technology measuring changes in myosin light chain phosphorylation can be used to rapidly generate quantitative IC(50) values and to screen a larger set of small molecule Rho-kinase inhibitors and suggests that this approach can be broadly applied to other cell lines and signaling pathways.


Subject(s)
Drug Evaluation, Preclinical/methods , Myosin Light Chains/analysis , Phosphoproteins/analysis , Protein Kinase Inhibitors/isolation & purification , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Cell Line , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology
15.
J Smooth Muscle Res ; 44(1): 29-40, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18480596

ABSTRACT

Myosin light chain genes of human hematopoietic cells have not been fully characterized. We previously reported the cloning of the full-length cDNAs of 20 kDa regulatory myosin light chain (MLC-2), named as MLC-2A, from Meg-01, a human megakaryoblastic leukemia cell line (J. Smooth Muscle Res. 37: 25-38, 2001). We now cloned another MLC-2 isoforms from human platelets and U937, a human monocytic leukemia cell line, named as MLC-2B and MLC-2C, respectively. Both MLC-2A and MLC-2B consisted of three exons, which were situated on gene loci 18p1.3. Analysis of the gene structure indicated that MLC-2A and MLC-2B utilized different exons. MLC-2C also consisted of three exons, which was situated on gene loci 20p12. Amino acid sequence of MLC-2C was, of interest, apparently almost the same as that of MLC-2 from chicken gizzard smooth muscle LC20-A (one amino acid's difference) and human vascular smooth muscle LC-20 (two amino acids' difference). All three protein kinase C phosphorylation residues (Ser-1, Ser-2, Thr-9) and both myosin light chain kinase phosporylation residues (Thr-18, Ser-19) are conserved in these three isoforms. The MLC-2A and MLC-2B mRNA were expressed constitutively in all of the human hematopoietic cell lines examined and their expression levels were almost the same. On the other hand, MLC-2C mRNA was expressed in untreated monocytic cell lines (U937 and A-THP-1) and HL-60 differentiated into monocyte/macrophage cell lineage by TPA treatment. These results indicate that smooth muscle type isoform, MLC-2C is the inducible isoform, and might play a crucial role in monocyte/macrophage cell lineage.


Subject(s)
Cardiac Myosins/metabolism , Macrophages/metabolism , Monocytes/metabolism , Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cardiac Myosins/analysis , Cardiac Myosins/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Chickens , Exons , Humans , Macrophages/cytology , Macrophages/pathology , Molecular Sequence Data , Monocytes/cytology , Monocytes/pathology , Myosin Light Chains/analysis , Myosin Light Chains/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism
16.
Acta Histochem ; 110(2): 172-7, 2008.
Article in English | MEDLINE | ID: mdl-17961638

ABSTRACT

The cytoskeleton of cerebral microvascular endothelial cells is a critical determinant of blood-brain barrier (BBB) function. Barrier integrity appears to be particularly sensitive to the phosphorylation state of specific residues within myosin regulatory light chain (RLC), one of two accessory light chains of the myosin II motor complex. Phosphorylation of myosin RLC by myosin light chain kinase (MLCK) has been implicated in BBB dysfunction associated with alcohol abuse and hypoxia, whereas dephosphorylation may enhance BBB integrity following exposure to lipid-lowering statin drugs. Using immunohistochemistry we provide evidence of widespread myosin II RLC distribution throughout the cerebral vasculature of the mouse. Light microscopy revealed immunolocalization of myosin II RLC protein in the endothelium of brain capillaries, the endothelial cell layer of arterioles and in association with venules. Immunolabeling of myosin RLC in non-muscle endothelial cells could be distinguished from myosin RLC immunoreactivity associated with the smooth muscle layer of the tunica media in larger muscular arterioles. These findings support an emerging role for myosin II RLC as a component of the actomyosin cytoskeleton of cerebral endothelial cells with the potential to contribute to the selective vulnerability of the brain in vivo.


Subject(s)
Brain/blood supply , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Animals , Cerebral Aqueduct/cytology , Cerebral Aqueduct/metabolism , Ependyma/cytology , Ependyma/metabolism , Immune Sera , Immunohistochemistry , Male , Mice , Microcirculation/cytology , Myosin Light Chains/analysis , Myosin Type II/analysis
17.
Stem Cell Reports ; 11(3): 828-841, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30122443

ABSTRACT

To facilitate understanding of human cardiomyocyte (CM) subtype specification, and the study of ventricular CM biology in particular, we developed a broadly applicable strategy for enrichment of ventricular cardiomyocytes (VCMs) derived from human embryonic stem cells (hESCs). A bacterial artificial chromosome transgenic H9 hESC line in which GFP expression was driven by the human ventricular-specific myosin light chain 2 (MYL2) promoter was generated, and screened to identify cell-surface markers specific for MYL2-GFP-expressing VCMs. A CD77+/CD200- cell-surface signature facilitated isolation of >97% cardiac troponin I-positive cells from H9 hESC differentiation cultures, with 65% expressing MYL2-GFP. This study provides a tool for VCM enrichment when using some, but not all, human pluripotent stem cell lines. Tools generated in this study can be utilized toward understanding CM subtype specification, and enriching for VCMs for therapeutic applications.


Subject(s)
Heart Ventricles/cytology , Human Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Antigens, CD/analysis , Cardiac Myosins/analysis , Cell Differentiation , Cell Line , Cells, Cultured , Humans , Myosin Light Chains/analysis , Trihexosylceramides/analysis
18.
Cardiovasc Pathol ; 34: 50-57, 2018.
Article in English | MEDLINE | ID: mdl-29677652

ABSTRACT

Skeletal myocytes have well-established fast and slow twitch fibers with unique gene and protein specific expression patterns. By immunohistochemical staining, these show a mosaic pattern across myocytes. We hypothesized cardiac myocytes may behave similarly where some proteins are differentially expressed between mature cardiomyocytes. We utilized the tool HPASubC on over 52,000 cardiac images of the Human Protein Atlas to identify differential protein expression patterns by immunohistochemistry across the cardiomyocytes. We matched identified proteins to open chromatin and gene expression data. We identified 143 putative proteins with mosaic patterns of expression across the cardiomyocytes. We validated four of these proteins (MYL3, MYL4, PAM, and MYOM1) and demonstrated unique atrial or ventricular patterns of expression for each. Acetylation of histone H3K27 at the promoters of these four genes were consistent with the atrial/ventricular expression patterns. Despite the generally accepted homogeneity of cardiomyocytes, a small subset of proteins varies between cardiomyocytes in a mosaic pattern. This fundamental process has been previously uncharacterized. These changes may inform on different functional and disease-related activities of proteins in individual cardiomyocytes.


Subject(s)
Muscle Proteins/analysis , Myocytes, Cardiac/chemistry , Acetylation , Amidine-Lyases/analysis , Connectin/analysis , Gene Expression Regulation , Histones/chemistry , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Mixed Function Oxygenases/analysis , Mosaicism , Muscle Proteins/genetics , Myosin Light Chains/analysis , Pattern Recognition, Automated , Phenotype , Promoter Regions, Genetic , Protein Interaction Maps , Proteomics/methods
19.
Mol Biol Cell ; 15(7): 3497-508, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15133124

ABSTRACT

To study the dynamics of stress fiber components in cultured fibroblasts, we expressed alpha-actinin and the myosin II regulatory myosin light chain (MLC) as fusion proteins with green fluorescent protein. Myosin activation was stimulated by treatment with calyculin A, a serine/threonine phosphatase inhibitor that elevates MLC phosphorylation, or with LPA, another agent that ultimately stimulates phosphorylation of MLC via a RhoA-mediated pathway. The resulting contraction caused stress fiber shortening and allowed observation of changes in the spacing of stress fiber components. We have observed that stress fibers, unlike muscle myofibrils, do not contract uniformly along their lengths. Although peripheral regions shortened, more central regions stretched. We detected higher levels of MLC and phosphorylated MLC in the peripheral region of stress fibers. Fluorescence recovery after photobleaching revealed more rapid exchange of myosin and alpha-actinin in the middle of stress fibers, compared with the periphery. Surprisingly, the widths of the myosin and alpha-actinin bands in stress fibers also varied in different regions. In the periphery, the banding patterns for both proteins were shorter, whereas in central regions, where stretching occurred, the bands were wider.


Subject(s)
Actinin/metabolism , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Stress Fibers/physiology , Stress Fibers/ultrastructure , Actinin/analysis , Actinin/genetics , Animals , Fibroblasts/immunology , Fibroblasts/metabolism , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Lysophospholipids/pharmacology , Marine Toxins , Mice , Myosin Light Chains/analysis , Myosin Light Chains/genetics , Myosin Type II/analysis , Myosin Type II/genetics , Oxazoles/pharmacology , Phosphorylation/drug effects , Photobleaching , Swiss 3T3 Cells
20.
Oncotarget ; 7(44): 71390-71399, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27655687

ABSTRACT

Rectal cancer treatment still fails with local and distant relapses of the disease. It is hypothesized that radiotherapy could stimulate cancer cell dissemination and metastasis. In this study, we evaluated the effect of X-radiation on collagen type I strap formation potential, i.e. matrix remodeling associated with mesenchymal cell migration, and behaviors of SW480, SW620, HCT116 p53+/+ and HCT116 p53-/- colon cancer cells. We determined a radiation-induced increase in collagen type I strap formation and migration potentials of SW480 and HCT116 p53+/+. Further studies with HCT116 p53+/+, indicated that after X-radiation strap forming cells have an increased motility. More, we detected a decrease in adhesion potential and mature integrin ß1 expression, but no change in non-muscle myosin II expression for HCT116 p53+/+ after X-radiation. Integrin ß1 neutralization resulted in a decreased cell adhesion and collagen type I strap formation in both sham and X-radiated conditions. Our study indicates collagen type I strap formation as a potential mechanism of colon cancer cells with increased migration potential after X-radiation, and suggests that other molecules than integrin ß1 and non-muscle myosin II are responsible for the radiation-induced collagen type I strap formation potential of colon cancer cells. This work encourages further molecular investigation of radiation-induced migration to improve rectal cancer treatment outcome.


Subject(s)
Collagen Type I/chemistry , Colonic Neoplasms/pathology , Cardiac Myosins/analysis , Cell Adhesion/radiation effects , Cell Line, Tumor , Cell Movement/radiation effects , Humans , Integrin beta1/physiology , Molecular Motor Proteins/analysis , Molecular Motor Proteins/physiology , Myosin Heavy Chains/analysis , Myosin Heavy Chains/physiology , Myosin Light Chains/analysis , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL