Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
1.
Hum Mol Genet ; 29(24): 3882-3891, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33355670

ABSTRACT

Striated preferentially expressed gene (SPEG), a member of the myosin light chain kinase family, is localized at the level of triad surrounding myofibrils in skeletal muscles. In humans, SPEG mutations are associated with centronuclear myopathy and cardiomyopathy. Using a striated muscle-specific Speg-knockout (KO) mouse model, we have previously shown that SPEG is critical for triad maintenance and calcium handling. Here, we further examined the molecular function of SPEG and characterized the effects of SPEG deficiency on triad and focal adhesion proteins. We used yeast two-hybrid assay, and identified desmin, an intermediate filament protein, to interact with SPEG and confirmed this interaction by co-immunoprecipitation. Using domain-mapping assay, we defined that Ig-like and fibronectin III domains of SPEG interact with rod domain of desmin. In skeletal muscles, SPEG depletion leads to desmin aggregates in vivo and a shift in desmin equilibrium from soluble to insoluble fraction. We also profiled the expression and localization of triadic proteins in Speg-KO mice using western blot and immunofluorescence. The amount of RyR1 and triadin were markedly reduced, whereas DHPRα1, SERCA1 and triadin were abnormally accumulated in discrete areas of Speg-KO myofibers. In addition, Speg-KO muscles exhibited internalized vinculin and ß1 integrin, both of which are critical components of the focal adhesion complex. Further, ß1 integrin was abnormally accumulated in early endosomes of Speg-KO myofibers. These results demonstrate that SPEG-deficient skeletal muscles exhibit several pathological features similar to those seen in MTM1 deficiency. Defects of shared cellular pathways may underlie these structural and functional abnormalities in both types of diseases.


Subject(s)
Cell Adhesion Molecules/metabolism , Desmin/metabolism , Focal Adhesions/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/pathology , Myosin-Light-Chain Kinase/physiology , Animals , Calcium/metabolism , Cell Adhesion Molecules/genetics , Desmin/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Mutation , Myopathies, Structural, Congenital/etiology , Myopathies, Structural, Congenital/metabolism
2.
Dev Biol ; 463(1): 88-98, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32361004

ABSTRACT

In adult Hydra, epitheliomuscle cells form the monolayered ecto- and endodermal epithelia. Their basal myonemes function as a longitudinal and circular muscle, respectively. Based on the observation that a Rho/Rock pathway, controlling the cell shape changes during detachment of Hydra buds, is not involved in body movement, at least two actomyosin compartments must exist in these cells: a basal one for body movement and a cortical one for cell shape changes. We therefore analyzed the regional and subcellular localization of the Ser19-phosphorylated myosin regulatory light chain (pMLC20). Along the body column, pMLC20 was detected strongly in the basal myonemes and weakly in the apical cell compartments of ectodermal epitheliomuscle cells. In cells of the bud base undergoing morphogenesis, pMLC20 was localized to intracellular stress fibers as well as to the apical and additionally to the lateral cortical compartment. Pharmacological inhibition revealed that pMLC20 is induced in these compartments by at least two independent pathways. In myonemes, MLC is phosphorylated mainly by myosin light chain kinase (MLCK). In contrast, the cortical apical and lateral MLC phosphorylation in constricting ectodermal cells of the bud base is stimulated via the Rho/ROCK pathway.


Subject(s)
Actomyosin/metabolism , Muscle Contraction/physiology , Myosin Light Chains/metabolism , Actin Cytoskeleton/metabolism , Actomyosin/physiology , Animals , Cell Shape , Epithelial Cells/metabolism , Hydra/metabolism , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Kinase/physiology , Phosphorylation , Signal Transduction , Stress Fibers/metabolism , rho-Associated Kinases/metabolism
3.
Int J Mol Sci ; 21(10)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443411

ABSTRACT

The intestinal epithelial apical junctional complex, which includes tight and adherens junctions, contributes to the intestinal barrier function via their role in regulating paracellular permeability. Myosin light chain II (MLC-2), has been shown to be a critical regulatory protein in altering paracellular permeability during gastrointestinal disorders. Previous studies have demonstrated that phosphorylation of MLC-2 is a biochemical marker for perijunctional actomyosin ring contraction, which increases paracellular permeability by regulating the apical junctional complex. The phosphorylation of MLC-2 is dominantly regulated by myosin light chain kinase- (MLCK-) and Rho-associated coiled-coil containing protein kinase- (ROCK-) mediated pathways. In this review, we aim to summarize the current state of knowledge regarding the role of MLCK- and ROCK-mediated pathways in the regulation of the intestinal barrier during normal homeostasis and digestive diseases. Additionally, we will also suggest potential therapeutic targeting of MLCK- and ROCK-associated pathways in gastrointestinal disorders that compromise the intestinal barrier.


Subject(s)
Intestinal Mucosa/physiology , Myosin-Light-Chain Kinase/metabolism , rho-Associated Kinases/metabolism , Animals , Humans , Intestinal Mucosa/metabolism , Myosin-Light-Chain Kinase/physiology , Permeability , Tight Junctions/metabolism , rho-Associated Kinases/physiology
4.
Am J Pathol ; 186(5): 1151-65, 2016 05.
Article in English | MEDLINE | ID: mdl-26948423

ABSTRACT

Tumor necrosis factor (TNF)-α, a key mediator of intestinal inflammation, causes an increase in intestinal epithelial tight junction (TJ) permeability by activating myosin light chain kinase (MLCK; official name MYLK3) gene. However, the precise signaling cascades that mediate the TNF-α-induced activation of MLCK gene and increase in TJ permeability remain unclear. Our aims were to delineate the upstream signaling mechanisms that regulate the TNF-α modulation of intestinal TJ barrier function with the use of in vitro and in vivo intestinal epithelial model systems. TNF-α caused a rapid activation of both canonical and noncanonical NF-κB pathway. NF-κB-inducing kinase (NIK) and mitogen-activated protein kinase kinase-1 (MEKK-1) were activated in response to TNF-α. NIK mediated the TNF-α activation of inhibitory κB kinase (IKK)-α, and MEKK1 mediated the activation of IKK complex, including IKK-ß. NIK/IKK-α axis regulated the activation of both NF-κB p50/p65 and RelB/p52 pathways. Surprisingly, the siRNA induced knockdown of NIK, but not MEKK-1, prevented the TNF-α activation of both NF-κB p50/p65 and RelB/p52 and the increase in intestinal TJ permeability. Moreover, NIK/IKK-α/NF-κB p50/p65 axis mediated the TNF-α-induced MLCK gene activation and the subsequent MLCK increase in intestinal TJ permeability. In conclusion, our data show that NIK/IKK-α/regulates the activation of NF-κB p50/p65 and plays an integral role in the TNF-α-induced activation of MLCK gene and increase in intestinal TJ permeability.


Subject(s)
I-kappa B Kinase/metabolism , MAP Kinase Kinase Kinase 1/metabolism , NF-kappa B/metabolism , Tight Junctions/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Caco-2 Cells , Cells, Cultured , Humans , Intestine, Small/physiology , MAP Kinase Signaling System/physiology , Mice , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Permeability , Promoter Regions, Genetic/physiology , RNA, Small Interfering/metabolism , Transcription Factor RelA/metabolism , Transfection , Tumor Necrosis Factor-alpha/pharmacology
5.
J Cell Sci ; 127(Pt 8): 1840-53, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24522189

ABSTRACT

Aberrant elevation in the levels of the pro-inflammatory cytokine interleukin-1ß (IL-1ß) contributes to neuroinflammatory diseases. Blood-brain barrier (BBB) dysfunction is a hallmark phenotype of neuroinflammation. It is known that IL-1ß directly induces BBB hyperpermeability but the mechanisms remain unclear. Claudin-5 (Cldn5) is a tight junction protein found at endothelial cell-cell contacts that are crucial for maintaining brain microvascular endothelial cell (BMVEC) integrity. Transcriptional regulation of Cldn5 has been attributed to the transcription factors ß-catenin and forkhead box protein O1 (FoxO1), and the signaling molecules regulating their nuclear translocation. Non-muscle myosin light chain kinase (nmMlck, encoded by the Mylk gene) is a key regulator involved in endothelial hyperpermeability, and IL-1ß has been shown to mediate nmMlck-dependent barrier dysfunction in epithelia. Considering these factors, we tested the hypothesis that nmMlck modulates IL-1ß-mediated downregulation of Cldn5 in BMVECs in a manner that depends on transcriptional repression mediated by ß-catenin and FoxO1. We found that treating BMVECs with IL-1ß induced barrier dysfunction concomitantly with the nuclear translocation of ß-catenin and FoxO1 and the repression of Cldn5. Most importantly, using primary BMVECs isolated from mice null for nmMlck, we identified that Cldn5 repression caused by ß-catenin and FoxO1 in IL-1ß-mediated barrier dysfunction was dependent on nmMlck.


Subject(s)
Blood-Brain Barrier/physiopathology , Claudin-5/genetics , Endothelial Cells/physiology , Forkhead Transcription Factors/physiology , Interleukin-1beta/physiology , Myosin-Light-Chain Kinase/physiology , beta Catenin/physiology , Animals , Antigens, CD/metabolism , Brain/blood supply , Cadherins/metabolism , Cells, Cultured , Claudin-5/metabolism , Down-Regulation , Endothelium, Vascular/physiopathology , Forkhead Box Protein O1 , Mice , Microvessels/pathology , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcriptional Activation
6.
Hepatology ; 61(3): 883-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25251280

ABSTRACT

UNLABELLED: Intestinal barrier dysfunction is an important contributor to alcoholic liver disease (ALD). Translocated microbial products trigger an inflammatory response in the liver and contribute to steatohepatitis. Our aim was to investigate mechanisms of barrier disruption after chronic alcohol feeding. A Lieber-DeCarli model was used to induce intestinal dysbiosis, increased intestinal permeability, and liver disease in mice. Alcohol feeding for 8 weeks induced intestinal inflammation in the jejunum, which is characterized by an increased number of tumor necrosis factor alpha (TNF-α)-producing monocytes and macrophages. These findings were confirmed in duodenal biopsies from patients with chronic alcohol abuse. Intestinal decontamination with nonabsorbable antibiotics restored eubiosis, decreased intestinal inflammation and permeability, and reduced ALD in mice. TNF-receptor I (TNFRI) mutant mice were protected from intestinal barrier dysfunction and ALD. To investigate whether TNFRI on intestinal epithelial cells mediates intestinal barrier dysfunction and ALD, we used TNFRI mutant mice carrying a conditional gain-of-function allele for this receptor. Reactivation of TNFRI on intestinal epithelial cells resulted in increased intestinal permeability and liver disease that is similar to wild-type mice after alcohol feeding, suggesting that enteric TNFRI promotes intestinal barrier dysfunction. Myosin light-chain kinase (MLCK) is a downstream target of TNF-α and was phosphorylated in intestinal epithelial cells after alcohol administration. Using MLCK-deficient mice, we further demonstrate a partial contribution of MLCK to intestinal barrier dysfunction and liver disease after chronic alcohol feeding. CONCLUSION: Dysbiosis-induced intestinal inflammation and TNFRI signaling in intestinal epithelial cells mediate a disruption of the intestinal barrier. Therefore, intestinal TNFRI is a crucial mediator of ALD.


Subject(s)
Dysbiosis/complications , Liver Diseases, Alcoholic/etiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Animals , Enteritis/complications , Intestinal Mucosa/physiology , Jejunal Diseases/complications , Mice , Mice, Inbred C57BL , Myosin-Light-Chain Kinase/physiology , Permeability , Tumor Necrosis Factor-alpha/genetics
7.
Biochemistry (Mosc) ; 81(13): 1676-1697, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28260490

ABSTRACT

This review discusses and summarizes the results of molecular and cellular investigations of myosin light chain kinase (MLCK, MYLK1), the key regulator of cell motility. The structure and regulation of a complex mylk1 gene and the domain organization of its products is presented. The interactions of the mylk1 gene protein products with other proteins and posttranslational modifications of the mylk1 gene protein products are reviewed, which altogether might determine the role and place of MLCK in physiological and pathological reactions of cells and entire organisms. Translational potential of MLCK as a drug target is evaluated.


Subject(s)
Calcium-Binding Proteins/physiology , Myosin-Light-Chain Kinase/physiology , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Gene Expression Regulation , Humans , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/genetics , Protein Domains , Protein Processing, Post-Translational
8.
J Mol Cell Cardiol ; 85: 199-206, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26057075

ABSTRACT

The effect of phosphorylation on the conformation of the regulatory light chain (cRLC) region of myosin in ventricular trabeculae from rat heart was determined by polarized fluorescence from thiophosphorylated cRLCs labelled with bifunctional sulforhodamine (BSR). Less than 5% of cRLCs were endogenously phosphorylated in this preparation, and similarly low values of basal cRLC phosphorylation were measured in fresh intact ventricle from both rat and mouse hearts. BSR-labelled cRLCs were thiophosphorylated by a recombinant fragment of human cardiac myosin light chain kinase, which was shown to phosphorylate cRLCs specifically at serine 15 in a calcium- and calmodulin-dependent manner, both in vitro and in situ. The BSR-cRLCs were exchanged into demembranated trabeculae, and polarized fluorescence intensities measured for each BSR-cRLC in relaxation, active isometric contraction and rigor were combined with RLC crystal structures to calculate the orientation distribution of the C-lobe of the cRLC in each state. Only two of the four C-lobe orientation populations seen during relaxation and active isometric contraction in the unphosphorylated state were present after cRLC phosphorylation. Thus cRLC phosphorylation alters the equilibrium between defined conformations of the cRLC regions of the myosin heads, rather than simply disordering the heads as assumed previously. cRLC phosphorylation also changes the orientation of the cRLC C-lobe in rigor conditions, showing that the orientation of this part of the myosin head is determined by its interaction with the thick filament even when the head is strongly bound to actin. These results suggest that cRLC phosphorylation controls the contractility of the heart by modulating the interaction of the cRLC region of the myosin heads with the thick filament backbone.


Subject(s)
Myosin Light Chains/chemistry , Protein Processing, Post-Translational , Animals , Humans , Myocardium/chemistry , Myocardium/metabolism , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/physiology , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Protein Conformation , Rats, Wistar
9.
J Biol Chem ; 289(32): 22512-23, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24951589

ABSTRACT

Myosin light chain phosphatase with its regulatory subunit, myosin phosphatase target subunit 1 (MYPT1) modulates Ca(2+)-dependent phosphorylation of myosin light chain by myosin light chain kinase, which is essential for smooth muscle contraction. The role of MYPT1 in vascular smooth muscle was investigated in adult MYPT1 smooth muscle specific knock-out mice. MYPT1 deletion enhanced phosphorylation of myosin regulatory light chain and contractile force in isolated mesenteric arteries treated with KCl and various vascular agonists. The contractile responses of arteries from knock-out mice to norepinephrine were inhibited by Rho-associated kinase (ROCK) and protein kinase C inhibitors and were associated with inhibition of phosphorylation of the myosin light chain phosphatase inhibitor CPI-17. Additionally, stimulation of the NO/cGMP/protein kinase G (PKG) signaling pathway still resulted in relaxation of MYPT1-deficient mesenteric arteries, indicating phosphorylation of MYPT1 by PKG is not a major contributor to the relaxation response. Thus, MYPT1 enhances myosin light chain phosphatase activity sufficient for blood pressure maintenance. Rho-associated kinase phosphorylation of CPI-17 plays a significant role in enhancing vascular contractile responses, whereas phosphorylation of MYPT1 in the NO/cGMP/PKG signaling module is not necessary for relaxation.


Subject(s)
Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Kinase/physiology , Animals , Blood Pressure/physiology , Female , Hypertension/etiology , Hypertension/physiopathology , Intracellular Signaling Peptides and Proteins , Male , Mesenteric Arteries/physiology , Mice , Mice, Knockout , Muscle Proteins/metabolism , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/deficiency , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Phosphatase , Nitric Oxide/metabolism , Phosphoproteins/metabolism , Phosphorylation , Signal Transduction , Vasoconstriction/physiology , Vasodilation/physiology
10.
EMBO J ; 30(4): 636-51, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21169990

ABSTRACT

Autophagy is a membrane-mediated degradation process of macromolecule recycling. Although the formation of double-membrane degradation vesicles (autophagosomes) is known to have a central role in autophagy, the mechanism underlying this process remains elusive. The serine/threonine kinase Atg1 has a key role in the induction of autophagy. In this study, we show that overexpression of Drosophila Atg1 promotes the phosphorylation-dependent activation of the actin-associated motor protein myosin II. A novel myosin light chain kinase (MLCK)-like protein, Spaghetti-squash activator (Sqa), was identified as a link between Atg1 and actomyosin activation. Sqa interacts with Atg1 through its kinase domain and is a substrate of Atg1. Significantly, myosin II inhibition or depletion of Sqa compromised the formation of autophagosomes under starvation conditions. In mammalian cells, we found that the Sqa mammalian homologue zipper-interacting protein kinase (ZIPK) and myosin II had a critical role in the regulation of starvation-induced autophagy and mammalian Atg9 (mAtg9) trafficking when cells were deprived of nutrients. Our findings provide evidence of a link between Atg1 and the control of Atg9-mediated autophagosome formation through the myosin II motor protein.


Subject(s)
Autophagy/physiology , Drosophila Proteins/physiology , Myosin Type II/metabolism , Phagosomes/metabolism , Protein Serine-Threonine Kinases/physiology , Starvation/metabolism , Animals , Animals, Genetically Modified , Autophagy/genetics , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Cells, Cultured , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Kinase/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Starvation/genetics , Tissue Distribution
11.
EMBO J ; 30(12): 2477-89, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21556048

ABSTRACT

The MEF2 factors regulate transcription during cardiac and skeletal myogenesis. MEF2 factors establish skeletal muscle commitment by amplifying and synergizing with MyoD. While phosphorylation is known to regulate MEF2 function, lineage-specific regulation is unknown. Here, we show that phosphorylation of MEF2C on T(80) by skeletal myosin light chain kinase (skMLCK) enhances skeletal and not cardiac myogenesis. A phosphorylation-deficient MEF2C mutant (MEFT80A) enhanced cardiac, but not skeletal myogenesis in P19 stem cells. Further, MEFT80A was deficient in recruitment of p300 to skeletal but not cardiac muscle promoters. In gain-of-function studies, skMLCK upregulated myogenic regulatory factor (MRF) expression, leading to enhanced skeletal myogenesis in P19 cells and more efficient myogenic conversion. In loss-of-function studies, MLCK was essential for efficient MRF expression and subsequent myogenesis in embryonic stem (ES) and P19 cells as well as for proper activation of quiescent satellite cells. Thus, skMLCK regulates MRF expression by controlling the MEF2C-dependent recruitment of histone acetyltransferases to skeletal muscle promoters. This work identifies the first kinase that regulates MyoD and Myf5 expression in ES or satellite cells.


Subject(s)
MADS Domain Proteins/metabolism , Muscle Development/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Myogenic Regulatory Factors/metabolism , Myosin-Light-Chain Kinase/physiology , Amino Acid Sequence , Animals , Carcinoma, Embryonal/enzymology , Carcinoma, Embryonal/pathology , Cell Line, Tumor , HEK293 Cells , Humans , MADS Domain Proteins/physiology , MEF2 Transcription Factors , Mice , Molecular Sequence Data , Myogenic Regulatory Factors/physiology , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Organ Specificity , Phosphorylation , Threonine/metabolism
12.
Biol Reprod ; 92(4): 97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25761595

ABSTRACT

The mammalian oocyte undergoes two rounds of asymmetric cell divisions during meiotic maturation and fertilization. Acentric spindle positioning and cortical polarity are two major factors involved in asymmetric cell division, both of which are thought to depend on the dynamic interaction between myosin II and actin filaments. Myosin light chain kinase (MLCK), encoded by the Mylk1 gene, could directly phosphorylate and activate myosin II. To determine whether MLCK was required for oocyte asymmetric division, we specifically disrupted the Mylk1 gene in oocytes by Cre-loxP conditional knockout system. We found that Mylk1 mutant female mice showed severe subfertility. Unexpectedly, contrary to previously reported in vitro findings, our data showed that oocyte meiotic maturation including spindle organization, polarity establishment, homologous chromosomes separation, and polar body extrusion were not affected in Mylk1(fl/fl);GCre(+) females. Follicular development, ovulation, and early embryonic development up to compact morula occurred normally in Mylk1(fl/fl);GCre(+) females, but deletion of MLCK caused delayed morula-to-blastocyst transition. More than a third of embryos were at morula stage at 3.5 Days Postcoitum in vivo. The delayed embryos could develop further to early blastocyst stage in vitro on Day 4 when most control embryos reached expanded blastocysts. Our findings provide evidence that MLCK is linked to timely blastocyst formation, though it is dispensable for oocyte meiotic maturation.


Subject(s)
Blastocyst/physiology , Fertility/genetics , Morula/physiology , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/physiology , Oocytes/physiology , Ovarian Follicle/physiology , Animals , Chromosomes, Mammalian/genetics , Female , Fertility/physiology , Fertilization/genetics , Gene Deletion , Infertility/genetics , Infertility/physiopathology , Meiosis/genetics , Mice , Mice, Inbred C57BL , Polar Bodies/physiology , Pregnancy , Spindle Apparatus/genetics , Spindle Apparatus/physiology
13.
J Pharmacol Sci ; 128(2): 78-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25986486

ABSTRACT

The role of myosin light chain kinase (MLCK) in inducing podosomes was examined by confocal and electron microscopy. Removal of myosin from the actin core of podosomes using blebbistatin, a myosin inhibitor, resulted in the formation of smaller podosomes. Downregulation of MLCK by the transfection of MLCK small interfering RNA (siRNA) led to the failure of podosome formation. However, ML-7, an inhibitor of the kinase activity of MLCK, failed to inhibit podosome formation. Based on our previous report (Thatcher et al. J.Pharm.Sci. 116 116-127, 2011), we outlined the important role of the actin-binding activity of MLCK in producing smaller podosomes.


Subject(s)
Myosin-Light-Chain Kinase/physiology , Phorbol 12,13-Dibutyrate/pharmacology , Podosomes/drug effects , Podosomes/ultrastructure , Actins/metabolism , Animals , Azepines/pharmacology , Cells, Cultured , Down-Regulation , Microscopy, Immunoelectron , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/pharmacology , Podosomes/genetics , Protein Binding , RNA, Small Interfering , Rats
14.
Proc Natl Acad Sci U S A ; 109(27): 10891-6, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22711834

ABSTRACT

Fundamental biological processes such as morphogenesis and wound healing involve the closure of epithelial gaps. Epithelial gap closure is commonly attributed either to the purse-string contraction of an intercellular actomyosin cable or to active cell migration, but the relative contribution of these two mechanisms remains unknown. Here we present a model experiment to systematically study epithelial closure in the absence of cell injury. We developed a pillar stencil approach to create well-defined gaps in terms of size and shape within an epithelial cell monolayer. Upon pillar removal, cells actively respond to the newly accessible free space by extending lamellipodia and migrating into the gap. The decrease of gap area over time is strikingly linear and shows two different regimes depending on the size of the gap. In large gaps, closure is dominated by lamellipodium-mediated cell migration. By contrast, closure of gaps smaller than 20 µm was affected by cell density and progressed independently of Rac, myosin light chain kinase, and Rho kinase, suggesting a passive physical mechanism. By changing the shape of the gap, we observed that low-curvature areas favored the appearance of lamellipodia, promoting faster closure. Altogether, our results reveal that the closure of epithelial gaps in the absence of cell injury is governed by the collective migration of cells through the activation of lamellipodium protrusion.


Subject(s)
Cell Movement/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Pseudopodia/physiology , Wound Healing/physiology , Actomyosin/physiology , Animals , Cell Count , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Line , Dogs , Intercellular Junctions/physiology , Kidney/cytology , Myosin-Light-Chain Kinase/physiology , Stress, Mechanical , rho-Associated Kinases/physiology
15.
J Physiol ; 592(14): 3031-51, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24835173

ABSTRACT

Smooth muscle contraction initiated by myosin regulatory light chain (RLC) phosphorylation is dependent on the relative activities of Ca(2+)-calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP). We have investigated the physiological role of the MLCP regulatory subunit MYPT1 in bladder smooth muscle containing a smooth muscle-specific deletion of MYPT1 in adult mice. Deep-sequencing analyses of mRNA and immunoblotting revealed that MYPT1 depletion reduced the amount of PP1cδ with no compensatory changes in expression of other MYPT1 family members. Phosphatase activity towards phosphorylated smooth muscle heavy meromyosin was proportional to the amount of PP1cδ in total homogenates from wild-type or MYPT1-deficient tissues. Isolated MYPT1-deficient tissues from MYPT1(SM-/-) mice contracted with moderate differences in response to KCl and carbachol treatments, and relaxed rapidly with comparable rates after carbachol removal and only 1.5-fold slower after KCl removal. Measurements of phosphorylated proteins in the RLC signalling and actin polymerization modules during contractions revealed moderate changes. Using a novel procedure to quantify total phosphorylation of MYPT1 at Thr696 and Thr853, we found substantial phosphorylation in wild-type tissues under resting conditions, predicting attenuation of MLCP activity. Reduced PP1cδ activity in MYPT1-deficient tissues may be similar to the attenuated MLCP activity in wild-type tissues resulting from constitutively phosphorylated MYPT1. Constitutive phosphorylation of MYPT1 Thr696 and Thr853 may thus represent a physiological mechanism acting in concert with agonist-induced MYPT1 phosphorylation to inhibit MLCP activity. In summary, MYPT1 deficiency may not cause significant derangement of smooth muscle contractility because the effective MLCP activity is not changed.


Subject(s)
Muscle, Smooth/physiology , Myosin-Light-Chain Kinase/physiology , Myosin-Light-Chain Phosphatase/physiology , Urinary Bladder/physiology , Animals , High-Throughput Nucleotide Sequencing , Male , Mice, Transgenic , Muscle Contraction , Phosphorylation , RNA, Messenger/genetics
16.
Gastroenterology ; 144(7): 1456-65, 1465.e1-5, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23499953

ABSTRACT

BACKGROUND & AIMS: The regulatory subunit of myosin light chain phosphatase, MYPT1, has been proposed to control smooth muscle contractility by regulating phosphorylation of the Ca(2+)-dependent myosin regulatory light chain. We generated mice with a smooth muscle-specific deletion of MYPT1 to investigate its physiologic role in intestinal smooth muscle contraction. METHODS: We used the Cre-loxP system to establish Mypt1-floxed mice, with the promoter region and exon 1 of Mypt1 flanked by 2 loxP sites. These mice were crossed with SMA-Cre transgenic mice to generate mice with smooth muscle-specific deletion of MYPT1 (Mypt1(SMKO) mice). The phenotype was assessed by histologic, biochemical, molecular, and physiologic analyses. RESULTS: Young adult Mypt1(SMKO) mice had normal intestinal motility in vivo, with no histologic abnormalities. On stimulation with KCl or acetylcholine, intestinal smooth muscles isolated from Mypt1(SMKO) mice produced robust and increased sustained force due to increased phosphorylation of the myosin regulatory light chain compared with muscle from control mice. Additional analyses of contractile properties showed reduced rates of force development and relaxation, and decreased shortening velocity, compared with muscle from control mice. Permeable smooth muscle fibers from Mypt1(SMKO) mice had increased sensitivity and contraction in response to Ca(2+). CONCLUSIONS: MYPT1 is not essential for smooth muscle function in mice but regulates the Ca(2+) sensitivity of force development and contributes to intestinal phasic contractile phenotype. Altered contractile responses in isolated tissues could be compensated by adaptive physiologic responses in vivo, where gut motility is affected by lower intensities of smooth muscle stimulation for myosin phosphorylation and force development.


Subject(s)
Calcium Signaling/physiology , Gastrointestinal Motility/physiology , Intestines/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Myosin-Light-Chain Kinase/physiology , Animals , Calcium/metabolism , Calcium Signaling/genetics , Female , Gastrointestinal Motility/genetics , Male , Mice , Mice, Knockout , Muscle Contraction/genetics , Myosin-Light-Chain Kinase/deficiency , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Phosphatase
17.
Gastroenterology ; 145(2): 407-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23619146

ABSTRACT

BACKGROUND & AIMS: Tight junction dysregulation and epithelial damage contribute to barrier loss in patients with inflammatory bowel disease. However, the mechanisms that regulate these processes and their relative contributions to disease pathogenesis are not completely understood. We investigated these processes using colitis models in mice. METHODS: We induced colitis by adoptive transfer of CD4(+)CD45RB(hi) cells or administration of dextran sulfate sodium to mice, including those deficient in tumor necrosis factor receptor (TNFR) 1, TNFR2, or the long isoform of myosin light chain kinase (MLCK). Intestinal tissues and isolated epithelial cells were analyzed by immunoblot, immunofluorescence, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction assays. RESULTS: Induction of immune-mediated colitis by CD4(+)CD45RB(hi) adoptive transfer increased intestinal permeability, epithelial expression of claudin-2, the long isoform of MLCK, and TNFR2 (but not TNFR1) and phosphorylation of the myosin II light chain. Long MLCK upregulation, myosin II light chain phosphorylation, barrier loss, and weight loss were attenuated in TNFR2(-/-) , but not TNFR1(-/-) , recipients of wild-type CD4(+)CD45RB(hi) cells. Similarly, long MLCK(-/-) mice had limited increases in myosin II light chain phosphorylation, claudin-2 expression, and intestinal permeability and delayed onset of adoptive transfer-induced colitis. However, coincident with onset of epithelial apoptosis, long MLCK(-/-) mice ultimately developed colitis. This indicates that disease progresses via apoptosis in the absence of MLCK-dependent tight junction regulation. In support of this conclusion, long MLCK(-/-) mice were not protected from epithelial apoptosis-mediated, damage-dependent dextran sulfate sodium colitis. CONCLUSIONS: In immune-mediated inflammatory bowel disease models, TNFR2 signaling increases long MLCK expression, resulting in tight junction dysregulation, barrier loss, and induction of colitis. At advanced stages, colitis progresses by apoptosis and mucosal damage that result in tight junction- and MLCK-independent barrier loss. Therefore, barrier loss in immune-mediated colitis occurs via two temporally and morphologically distinct mechanisms. Differential targeting of these mechanisms can lead to improved inflammatory bowel disease therapies.


Subject(s)
Colitis/physiopathology , Epithelial Cells/physiology , Intestinal Mucosa/physiopathology , Myosin-Light-Chain Kinase/physiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tight Junctions/physiology , Adoptive Transfer , Animals , Apoptosis/physiology , Claudin-2/metabolism , Colitis/immunology , Colitis/metabolism , Dextran Sulfate , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Inflammatory Bowel Diseases , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Myosin-Light-Chain Kinase/genetics , Permeability , Phosphorylation , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction , Tight Junctions/metabolism , Up-Regulation , Weight Loss
18.
J Surg Res ; 186(1): 304-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24075254

ABSTRACT

BACKGROUND: Vascular hyporeactivity plays an important role in the pathogenesis of severe shock. Previous studies have shown that postshock mesenteric lymph (PSML) blockage ameliorates the vascular reactivity and calcium sensitivity, and RhoA is involved in the regulation of vascular reactivity after hemorrhagic shock. Therefore, the present study tested whether small GTPase RhoA mediates the improvement of the vascular reactivity and calcium sensitivity in superior mesenteric artery (SMA) of rats with PSML drainage. MATERIALS AND METHODS: The hemorrhagic shock model (blood pressure to 40 ± 2 mm Hg) was established, and PSML was drained from immediate hypotension for 3 h, after which SMA was isolated, and the vascular reactivity and calcium sensitivity were tested in the presence of RhoA agonist (U-46619) or inhibitor (C3 transferase). The protein expressions of small GTPase RhoA and phospho-RhoA were also examined in SMA. RESULTS: The hemorrhagic shock resulted in a significant decrease in the SMA reactivity and calcium sensitivity, which was enhanced by the application of U-46619 to the SMA. In contrast, the PSML drainage ameliorated the deleterious effect of the hemorrhagic shock on the SMA. This beneficial effect of the PSML drainage was abolished by C3 transferase. Western blotting revealed that the expressions of the RhoA and phospho-RhoA in SMA tissue obtained from the shock group were significantly decreased, and the PSML drainage markedly enhanced these protein expressions. CONCLUSIONS: RhoA is an important contributor to the PSML drainage-induced amelioration of the vascular reactivity and calcium sensitivity in rats with hemorrhagic shock.


Subject(s)
Calcium/metabolism , Drainage , Lymph/physiology , Mesenteric Artery, Superior/physiopathology , Shock, Hemorrhagic/physiopathology , rhoA GTP-Binding Protein/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP Ribose Transferases/pharmacology , Animals , Botulinum Toxins/pharmacology , Male , Myosin-Light-Chain Kinase/physiology , Rats , Rats, Wistar , rhoA GTP-Binding Protein/antagonists & inhibitors
19.
J Physiol ; 591(15): 3709-23, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23753529

ABSTRACT

Tumour necrosis factor-α (TNF-α) has been reported to play a central role in intestinal barrier dysfunction in many diseases; however, the precise role of the TNF-α receptors (TNFRs) has not been well defined using in vivo models. Our previous data showed that enteral nutrient deprivation or total parenteral nutrition (TPN) led to a loss of intestinal epithelial barrier function (EBF), with an associated upregulation of TNF-α and TNFR1. In this study, we hypothesized that TNF-α plays an important role in TPN-associated EBF dysfunction. Using a mouse TPN model, we explored the relative roles of TNFR1 vs. TNFR2 in mediating this barrier loss. C57/BL6 mice underwent intravenous cannulation and were given enteral nutrition or TPN for 7 days. Tumour necrosis factor-α receptor knockout (KO) mice, including TNFR1KO, TNFR2KO or TNFR1R2 double KO (DKO), were used. Outcomes included small intestine transepithelial resistance (TER) and tracer permeability, junctional protein zonula occludens-1, occludin, claudins and E-cadherin expression. In order to address the dependence of EBF on TNF-α further, exogenous TNF-α and pharmacological blockade of TNF-α (Etanercept) were also performed. Total parenteral nutrition led to a loss of EBF, and this was almost completely prevented in TNFR1R2DKO mice and partly prevented in TNFR1KO mice but not in TNFR2KO mice. The TPN-associated downregulation of junctional protein expression and junctional assembly was almost completely prevented in the TNFR1R2DKO group. Blockade of TNF-α also prevented dysfunction of the EBF and junctional protein losses in mice undergoing TPN. Administration of TPN upregulated the downstream nuclear factor-B and myosin light-chain kinase (MLCK) signalling, and these changes were almost completely prevented in TNFR1R2DKO mice, as well as with TNF-α blockade, but not in TNFR1KO or TNFR2KO TPN groups. Tumour necrosis factor-α is a critical factor for TPN-associated epithelial barrier dysfunction, and both TNFR1 and TNFR2 are involved in EBF loss. Nuclear factor-B and MLCK signalling appear to be important downstream mediators involved in this TNF-α signalling process.


Subject(s)
Intestinal Mucosa/physiopathology , Parenteral Nutrition, Total/adverse effects , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Etanercept , Immunoglobulin G/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin-Light-Chain Kinase/physiology , NF-kappa B/physiology , Receptors, Tumor Necrosis Factor , Signal Transduction , Tight Junctions/metabolism
20.
Cell Mol Life Sci ; 69(2): 247-66, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21947498

ABSTRACT

Vascular smooth muscle tone is controlled by a balance between the cellular signaling pathways that mediate the generation of force (vasoconstriction) and release of force (vasodilation). The initiation of force is associated with increases in intracellular calcium concentrations, activation of myosin light-chain kinase, increases in the phosphorylation of the regulatory myosin light chains, and actin-myosin crossbridge cycling. There are, however, several signaling pathways modulating Ca(2+) mobilization and Ca(2+) sensitivity of the contractile machinery that secondarily regulate the contractile response of vascular smooth muscle to receptor agonists. Among these regulatory mechanisms involved in the physiological regulation of vascular tone are the cyclic nucleotides (cAMP and cGMP), which are considered the main messengers that mediate vasodilation under physiological conditions. At least four distinct mechanisms are currently thought to be involved in the vasodilator effect of cyclic nucleotides and their dependent protein kinases: (1) the decrease in cytosolic calcium concentration ([Ca(2+)]c), (2) the hyperpolarization of the smooth muscle cell membrane potential, (3) the reduction in the sensitivity of the contractile machinery by decreasing the [Ca(2+)]c sensitivity of myosin light-chain phosphorylation, and (4) the reduction in the sensitivity of the contractile machinery by uncoupling contraction from myosin light-chain phosphorylation. This review focuses on each of these mechanisms involved in cyclic nucleotide-dependent relaxation of vascular smooth muscle under physiological conditions.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Nucleotides, Cyclic/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Calcium/metabolism , Calcium/physiology , Calcium-Transporting ATPases/metabolism , Calcium-Transporting ATPases/physiology , Humans , Mice , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Kinase/physiology , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Myosin-Light-Chain Phosphatase/metabolism , Myosin-Light-Chain Phosphatase/physiology , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/physiology , Potassium Channels/agonists , Potassium Channels/metabolism , Potassium Channels/physiology , Rats , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/physiology , Sodium-Calcium Exchanger/metabolism , Sodium-Calcium Exchanger/physiology , Vasodilation/physiology , Vasodilator Agents/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL