Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(1): e0226138, 2020.
Article in English | MEDLINE | ID: mdl-31899774

ABSTRACT

A novel cardiac-specific transgenic mouse model was generated to identify the physiological consequences of elongated thin filaments during post-natal development in the heart. Remarkably, increasing the expression levels in vivo of just one sarcomeric protein, Lmod2, results in ~10% longer thin filaments (up to 26% longer in some individual sarcomeres) that produce up to 50% less contractile force. Increasing the levels of Lmod2 in vivo (Lmod2-TG) also allows us to probe the contribution of Lmod2 in the progression of cardiac myopathy because Lmod2-TG mice present with a unique cardiomyopathy involving enlarged atrial and ventricular lumens, increased heart mass, disorganized myofibrils and eventually, heart failure. Turning off of Lmod2 transgene expression at postnatal day 3 successfully prevents thin filament elongation, as well as gross morphological and functional disease progression. We show here that Lmod2 has an essential role in regulating cardiac contractile force and function.


Subject(s)
Actin Cytoskeleton/pathology , Cardiomyopathies/physiopathology , Cytoskeletal Proteins/physiology , Heart Failure/etiology , Muscle Proteins/physiology , Muscle, Skeletal/pathology , Sarcomeres/pathology , Animals , Animals, Newborn , Female , Heart Failure/pathology , Male , Mice , Mice, Transgenic , Muscle Contraction
2.
Circulation ; 137(6): 605-618, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29101288

ABSTRACT

BACKGROUND: Gap junction remodeling is well established as a consistent feature of human heart disease involving spontaneous ventricular arrhythmia. The mechanisms responsible for gap junction remodeling that include alterations in the distribution of, and protein expression within, gap junctions are still debated. Studies reveal that multiple transcriptional and posttranscriptional regulatory pathways are triggered in response to cardiac disease, such as those involving RNA-binding proteins. The expression levels of FXR1 (fragile X mental retardation autosomal homolog 1), an RNA-binding protein, are critical to maintain proper cardiac muscle function; however, the connection between FXR1 and disease is not clear. METHODS: To identify the mechanisms regulating gap junction remodeling in cardiac disease, we sought to identify the functional properties of FXR1 expression, direct targets of FXR1 in human left ventricle dilated cardiomyopathy (DCM) biopsy samples and mouse models of DCM through BioID proximity assay and RNA immunoprecipitation, how FXR1 regulates its targets through RNA stability and luciferase assays, and functional consequences of altering the levels of this important RNA-binding protein through the analysis of cardiac-specific FXR1 knockout mice and mice injected with 3xMyc-FXR1 adeno-associated virus. RESULTS: FXR1 expression is significantly increased in tissue samples from human and mouse models of DCM via Western blot analysis. FXR1 associates with intercalated discs, and integral gap junction proteins Cx43 (connexin 43), Cx45 (connexin 45), and ZO-1 (zonula occludens-1) were identified as novel mRNA targets of FXR1 by using a BioID proximity assay and RNA immunoprecipitation. Our findings show that FXR1 is a multifunctional protein involved in translational regulation and stabilization of its mRNA targets in heart muscle. In addition, introduction of 3xMyc-FXR1 via adeno-associated virus into mice leads to the redistribution of gap junctions and promotes ventricular tachycardia, showing the functional significance of FXR1 upregulation observed in DCM. CONCLUSIONS: In DCM, increased FXR1 expression appears to play an important role in disease progression by regulating gap junction remodeling. Together this study provides a novel function of FXR1, namely, that it directly regulates major gap junction components, contributing to proper cell-cell communication in the heart.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Gap Junctions/metabolism , Heart Ventricles/metabolism , RNA-Binding Proteins/metabolism , Tachycardia, Ventricular/etiology , Ventricular Function, Left , Ventricular Remodeling , Animals , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Communication , Connexins/genetics , Connexins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Gap Junctions/pathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , RNA-Binding Proteins/genetics , Rats , Risk Factors , Signal Transduction , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Up-Regulation , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
3.
J Exp Biol ; 219(Pt 2): 146-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26792324

ABSTRACT

Efficient muscle contraction in skeletal muscle is predicated on the regulation of actin filament lengths. In one long-standing model that was prominent for decades, the giant protein nebulin was proposed to function as a 'molecular ruler' to specify the lengths of the thin filaments. This theory was questioned by many observations, including experiments in which the length of nebulin was manipulated in skeletal myocytes; this approach revealed that nebulin functions to stabilize filamentous actin, allowing thin filaments to reach mature lengths. In addition, more recent data, mostly from in vivo models and identification of new interacting partners, have provided evidence that nebulin is not merely a structural protein. Nebulin plays a role in numerous cellular processes including regulation of muscle contraction, Z-disc formation, and myofibril organization and assembly.


Subject(s)
Muscle Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Disease , Humans , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutation/genetics , Protein Binding
4.
Proc Natl Acad Sci U S A ; 112(44): 13573-8, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26487682

ABSTRACT

Leiomodin 2 (Lmod2) is an actin-binding protein that has been implicated in the regulation of striated muscle thin filament assembly; its physiological function has yet to be studied. We found that knockout of Lmod2 in mice results in abnormally short thin filaments in the heart. We also discovered that Lmod2 functions to elongate thin filaments by promoting actin assembly and dynamics at thin filament pointed ends. Lmod2-KO mice die as juveniles with hearts displaying contractile dysfunction and ventricular chamber enlargement consistent with dilated cardiomyopathy. Lmod2-null cardiomyocytes produce less contractile force than wild type when plated on micropillar arrays. Introduction of GFP-Lmod2 via adeno-associated viral transduction elongates thin filaments and rescues structural and functional defects observed in Lmod2-KO mice, extending their lifespan to adulthood. Thus, to our knowledge, Lmod2 is the first identified mammalian protein that functions to elongate actin filaments in the heart; it is essential for cardiac thin filaments to reach a mature length and is required for efficient contractile force and proper heart function during development.


Subject(s)
Actin Cytoskeleton/metabolism , Cardiomyopathy, Dilated/metabolism , Cytoskeletal Proteins/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Actin Cytoskeleton/genetics , Animals , Animals, Newborn , Cardiomyopathy, Dilated/embryology , Cardiomyopathy, Dilated/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Fluorescence Recovery After Photobleaching , Genes, Lethal/genetics , Heart/embryology , Heart/physiopathology , Immunoblotting , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myocardium/pathology , Myocardium/ultrastructure , Sarcomeres/genetics , Sarcomeres/metabolism , Survival Analysis
5.
J Mol Cell Cardiol ; 72: 281-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24713463

ABSTRACT

Up-regulation and activation of PYK2, a member of the FAK family of protein tyrosine kinases, is involved in the pathogenesis of left ventricular (LV) remodeling and heart failure (HF). PYK2 activation can be prevented by CRNK, the C-terminal domain of PYK2. We previously demonstrated that adenoviral-mediated CRNK gene transfer improved survival and LV function, and slowed LV remodeling in a rat model of coronary artery ligation-induced HF. We now interrogate whether cardiomyocyte-specific, transgenic CRNK expression prevents LV remodeling and HF in a mouse model of dilated cardiomyopathy (DCM) caused by constitutively active Protein Kinase Cε (caPKCε). Transgenic (TG; FVB/N background) mice were engineered to express rat CRNK under control of the α-myosin heavy chain promoter, and crossed with FVB/N mice with cardiomyocyte-specific expression of caPKCε to create double TG mice. LV structure, function, and gene expression were evaluated in all 4 groups (nonTG FVB/N; caPKCε(+/-); CRNK(+/-); and caPKCε×CRNK (PXC) double TG mice) at 1, 3, 6, 9 and 12mo of age. CRNK expression followed a Mendelian distribution, and CRNK mice developed and survived normally through 12mo. Cardiac structure, function and selected gene expression of CRNK mice were similar to nonTG littermates. CRNK had no effect on caPKCε expression and vice versa. PYK2 was up-regulated ~6-fold in caPKCε mice, who developed a non-hypertrophic, progressive DCM with reduced systolic (Contractility Index=151±5 vs. 90±4s(-1)) and diastolic (Tau=7.5±0.5 vs. 14.7±1.3ms) function, and LV dilatation (LV Remodeling Index (LVRI)=4.2±0.1 vs. 6.0±0.3 for FVB/N vs. caPKCε mice, respectively; P<0.05 for each at 12mo). In double TG PXC mice, CRNK expression significantly prolonged survival, improved contractile function (Contractile Index=115±8s(-1); Tau=9.5±1.0ms), and reduced LV remodeling (LVRI=4.9±0.1). Cardiomyocyte-specific expression of CRNK improves contractile function and slows LV remodeling in a mouse model of DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , Focal Adhesion Kinase 2/genetics , Myocytes, Cardiac/metabolism , Transgenes , Ventricular Function/physiology , Ventricular Remodeling , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Focal Adhesion Kinase 2/deficiency , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/pathology , Longevity , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Promoter Regions, Genetic , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics , Protein Structure, Tertiary
6.
J Card Fail ; 19(4): 283-94, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23582094

ABSTRACT

BACKGROUND: Heart failure (HF) is associated with excessive extracellular matrix (ECM) deposition and abnormal ECM degradation leading to cardiac fibrosis. Connective tissue growth factor (CTGF) modulates ECM production during inflammatory tissue injury, but available data on CTGF gene expression in failing human heart and its response to mechanical unloading are limited. METHODS AND RESULTS: Left ventricle (LV) tissue from patients undergoing cardiac transplantation for ischemic (ICM; n = 20) and dilated (DCM; n = 20) cardiomyopathies and from nonfailing (NF; n = 20) donor hearts were examined. Paired samples (n = 15) from patients undergoing LV assist device (LVAD) implantation as "bridge to transplant" (34-1,145 days) also were analyzed. There was more interstitial fibrosis in both ICM and DCM compared with NF hearts. Hydroxyproline concentration was also significantly increased in DCM compared with NF samples. The expression of CTGF, transforming growth factor (TGF) ß1, collagen (COL) 1-α1, COL3-α1, matrix metalloproteinase (MMP) 2, and MMP9 mRNA in ICM and DCM were also significantly elevated compared with NF samples. Although TGF-ß1, CTGF, COL1-α1, and COL3-α1 mRNA levels were reduced by unloading, there was only a modest reduction in tissue fibrosis and no difference in protein-bound hydroxyproline concentration between pre- and post-LVAD tissue samples. The persistent fibrosis may be related to a concomitant reduction in MMP9 mRNA and protein levels following unloading. CONCLUSIONS: CTGF may be a key regulator of fibrosis during maladaptive remodeling and progression to HF. Although mechanical unloading normalizes most genotypic and functional abnormalities, its effect on ECM remodeling during HF is incomplete.


Subject(s)
Cardiomyopathy, Dilated/pathology , Connective Tissue Growth Factor/biosynthesis , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/pathology , Heart Transplantation , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/metabolism , Connective Tissue Growth Factor/genetics , Fibrosis , Heart Failure/diagnosis , Heart Transplantation/trends , Humans , Ventricular Remodeling/genetics
7.
Mol Biol Cell ; 24(7): 995-1006, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23389630

ABSTRACT

Focal adhesions are intricate protein complexes that facilitate cell attachment, migration, and cellular communication. Lasp-2 (LIM-nebulette), a member of the nebulin family of actin-binding proteins, is a newly identified component of these complexes. To gain further insights into the functional role of lasp-2, we identified two additional binding partners of lasp-2: the integral focal adhesion proteins vinculin and paxillin. Of interest, the interaction of lasp-2 with its binding partners vinculin and paxillin is significantly reduced in the presence of lasp-1, another nebulin family member. The presence of lasp-2 appears to enhance the interaction of vinculin and paxillin with each other; however, as with the interaction of lasp-2 with vinculin or paxillin, this effect is greatly diminished in the presence of excess lasp-1. This suggests that the interplay between lasp-2 and lasp-1 could be an adhesion regulatory mechanism. Lasp-2's potential role in metastasis is revealed, as overexpression of lasp-2 in either SW620 or PC-3B1 cells-metastatic cancer cell lines-increases cell migration but impedes cell invasion, suggesting that the enhanced interaction of vinculin and paxillin may functionally destabilize focal adhesion composition. Taken together, these data suggest that lasp-2 has an important role in coordinating and regulating the composition and dynamics of focal adhesions.


Subject(s)
Carrier Proteins/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Paxillin/metabolism , Vinculin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Carrier Proteins/genetics , Cell Adhesion , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , LIM Domain Proteins/genetics , Microscopy, Fluorescence , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques , Vinculin/genetics
8.
J Signal Transduct ; 2012: 473410, 2012.
Article in English | MEDLINE | ID: mdl-22900166

ABSTRACT

Intracellular nitric oxide (NO(i)) is a physiological regulator of excitation-contraction coupling, but is also involved in the development of cardiac dysfunction during hypertrophy and heart failure. To determine whether contractile activity regulates nitric oxide synthase (NOS) expression, spontaneously contracting, neonatal rat ventricular myocytes (NRVM) were treat with L-type calcium channel blockers (nifedipine and verapamil) or myosin II ATPase inhibitors (butanedione monoxime (BDM) and blebbistatin) to produce contractile arrest. Both types of inhibitors significantly reduced iNOS but not eNOS expression, and also reduced NO(i) production. Inhibiting contractile activity also reduced focal adhesion kinase (FAK) and AKT phosphorylation. Contraction-induced iNOS expression required FAK and phosphatidylinositol 3-kinase (PI(3)K), as both PF573228 and LY294002 (10 µM, 24 h) eliminated contraction-induced iNOS expression. Similarly, shRNAs specific for FAK (shFAK) caused FAK knockdown, reduced AKT phosphorylation at T308 and S473, and reduced iNOS expression. In contrast, shRNA-mediated knockdown of PYK2, the other member of the FAK-family of protein tyrosine kinases, had much less of an effect. Conversely, overexpression of a constitutively active form of FAK (CD2-FAK) or AKT (Myr-AKT) reversed the inhibitory effect of BDM on iNOS expression and NO(i) production. Thus, contraction-induced iNOS expression and NO(i) production in NRVM are mediated via a FAK-PI(3)K-AKT signaling pathway.

9.
Cardiovasc Res ; 92(3): 409-19, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21937583

ABSTRACT

AIMS: Tyrosine-phosphorylated focal adhesion kinase (FAK) is required for the hypertrophic response of cardiomyocytes to growth factors and mechanical load, but the role of FAK serine phosphorylation in this process is unknown. The aims of the present study were to characterize FAK serine phosphorylation in cultured neonatal rat ventricular myocytes (NRVM), analyse its functional significance during hypertrophic signalling, and examine its potential role in the pathogenesis of human dilated cardiomyopathy (DCM). METHODS AND RESULTS: Endothelin-1 (ET-1) and other hypertrophic factors induced a time- and dose-dependent increase in FAK-S910 phosphorylation. ET-1-induced FAK-S910 phosphorylation required ET(A)R-dependent activation of PKCδ and Src via parallel Raf-1 → MEK1/2 → ERK1/2 and MEK5 → ERK5 signalling pathways. Replication-deficient adenoviruses expressing wild-type (WT) FAK and a non-phosphorylatable, S910A-FAK mutant were then used to examine the functional significance of FAK-S910 phosphorylation. Unlike WT-FAK, S910A-FAK increased the half-life of GFP-tagged paxillin within costameres (as determined by total internal reflection fluorescence microscopy and fluorescence recovery after photobleaching) and increased the steady-state FAK-paxillin interaction (as determined by co-immunoprecipitation and western blotting). These alterations resulted in reduced NRVM sarcomere reorganization and cell spreading. Finally, we found that FAK was serine-phosphorylated at multiple sites in non-failing, human left ventricular tissue. FAK-S910 phosphorylation and ERK5 expression were dramatically reduced in patients undergoing heart transplantation for end-stage DCM. CONCLUSION: FAK undergoes S910 phosphorylation via PKCδ and Src-dependent pathways that are important for cell spreading and sarcomere reorganization. Reduced FAK-S910 phosphorylation may contribute to sarcomere disorganization in DCM.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Focal Adhesion Kinase 1/metabolism , Heart Failure/enzymology , Myocytes, Cardiac/enzymology , Sarcomeres/enzymology , Angiotensin II/pharmacology , Animals , Animals, Newborn , Blotting, Western , Cardiomyopathy, Dilated/pathology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelin-1/pharmacology , Enzyme Activation , Fluorescence Recovery After Photobleaching , Focal Adhesion Kinase 1/genetics , Heart Failure/pathology , Humans , Immunoprecipitation , Insulin-Like Growth Factor I/pharmacology , Microscopy, Fluorescence , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Paxillin/genetics , Paxillin/metabolism , Phenylephrine/pharmacology , Phosphorylation , Protein Kinase C-delta/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Sarcomeres/drug effects , Sarcomeres/pathology , Serine , Signal Transduction , Time Factors , Transfection , src-Family Kinases/metabolism
10.
Arterioscler Thromb Vasc Biol ; 31(11): 2432-40, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21852560

ABSTRACT

OBJECTIVE: Focal adhesion kinase-related nonkinase (FRNK), the C-terminal domain of focal adhesion kinase (FAK), is a tyrosine-phosphorylated, vascular smooth muscle cell (VSMC)-specific inhibitor of cell migration. FRNK inhibits both FAK and proline-rich tyrosine kinase 2 (PYK2) in cultured VSMCs, and both kinases may be involved in VSMC invasion during vascular remodeling. METHODS AND RESULTS: Adenovirally mediated gene transfer of green fluorescent protein-tagged, wild-type (wt) FRNK into balloon-injured rat carotid arteries confirmed that FRNK overexpression inhibited both FAK and PYK2 phosphorylation and downstream signaling in vivo. To identify which kinase was involved in regulating VSMC invasion, adenovirally mediated expression of specific short hairpin RNAs was used to knock down FAK versus PYK2 in cultured VSMCs, but only FAK short hairpin RNA was effective in reducing VSMC invasion. The role of FRNK tyrosine phosphorylation was then examined using adenoviruses expressing nonphosphorylatable (Tyr168Phe-, Tyr232Phe-, and Tyr168,232Phe-) green fluorescent protein-FRNK mutants. wtFRNK and all FRNK mutants localized to FAs, but only Tyr168 phosphorylation was required for FRNK to inhibit invasion. Preventing Tyr168 phosphorylation also increased FRNK-paxillin interaction, as determined by coimmunoprecipitation, total internal reflection fluorescence microscopy, and fluorescence recovery after photobleaching. Furthermore, wtFRNK competed with FAK for binding to p130(Cas) (a critically important regulator of cell migration) and prevented its phosphorylation. However, Tyr168Phe-FRNK was unable to bind p130(Cas). CONCLUSION: We propose a 3-stage mechanism for FRNK inhibition: focal adhesion targeting, Tyr168 phosphorylation, and competition with FAK for p130 binding and phosphorylation, which are all required for FRNK to inhibit VSMC invasion.


Subject(s)
Cell Movement/physiology , Crk-Associated Substrate Protein/metabolism , Focal Adhesion Kinase 2/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Protein-Tyrosine Kinases/metabolism , Adenoviridae/genetics , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Catheterization/adverse effects , Cells, Cultured , Focal Adhesion Kinase 2/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Models, Animal , Phosphorylation/physiology , Protein Binding/physiology , RNA, Small Interfering/pharmacology , Rats
11.
Arterioscler Thromb Vasc Biol ; 30(11): 2226-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20705914

ABSTRACT

OBJECTIVE: To examine whether interference with FRNK targeting to focal adhesions (FAs) affects its inhibitory activity and tyrosine phosphorylation. METHODS AND RESULTS: Focal adhesion kinase and its autonomously expressed C-terminal inhibitor, focal adhesion kinase-related nonkinase (FRNK), regulate vascular smooth muscle cell (VSMC) signaling and migration. FRNK-paxillin binding was reduced by a point mutation in its FA targeting domain (L341S-FRNK). Green fluorescent protein-tagged wild type and L341S-FRNK were then adenovirally expressed in VSMCs. L341S-FRNK targeted to VSMC FAs, despite previous studies in other cell types. L341S-FRNK affected FA binding kinetics (assessed by total internal reflection fluorescnece [TIRF] microscopy and fluorescence recovery after photobleaching [FRAP]) and reduced its steady-state paxillin interaction (determined by coimmunoprecipitation). Both wt-FRNK and L341S-FRNK lowered basal and angiotensin II-stimulated focal adhesion kinase, paxillin, and extracellular signal-regulated kinase 1/2 phosphorylation. However, the degree of inhibition was significantly reduced by L341S-FRNK. L341S-FRNK also demonstrated significantly greater migratory activity compared with wt-FRNK-expressing VSMCs. Angiotensin II-induced Y168 phosphorylation was Src dependent, as evident by a significant reduction in Y168 phosphorylation by the Src family kinase inhibitor PP2 is 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). Surprisingly, Y168 phosphorylation was unaffected by its targeting. Furthermore, Y232 phosphorylation increased approximately 3-fold in L341S-FRNK, which was less sensitive to PP2. CONCLUSIONS: FRNK inhibition of VSMC migration requires both FA targeting and Y168 phosphorylation by Src family kinases. FRNK-Y232 phosphorylation occurs outside of FAs, probably by a PP2-insensitive kinase.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Myocytes, Smooth Muscle/physiology , Paxillin/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Cell Movement/physiology , Muscle, Smooth, Vascular , Phosphorylation , Point Mutation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Rats , Signal Transduction
12.
Stem Cells Cloning ; 3: 165-73, 2010.
Article in English | MEDLINE | ID: mdl-24198522

ABSTRACT

The use of in vitro oligodendrocyte differentiation for transplantation of stem cells to treat demyelinating diseases is an important consideration. In this study, we investigated the effects of serum on glia and oligodendrocyte differentiation from human mesenchymal stem cells (KP-hMSCs). We found that serum deprivation resulted in a reversible downregulation of glial- and oligodendrocyte-specific markers. Serum stimulated expression of oligodendrocyte markers, such as galactocerebroside, as well as Notch1 and JAK1 transcripts. Inhibition of Notch1 activation by the Notch inhibitor, MG132, led to enhanced expression of a serum-stimulated oligodendrocyte marker. This marker was undetectable in serum-deprived KP-hMSCs treated with MG132, suggesting that inhibition of Notch1 function is additive to serum-stimulated oligodendrocyte differentiation. Furthermore, a dominant-negative mutant RBP-J protein also inhibited Notch1 function and led to upregulation of oligodendrocyte-specific markers. Our results demonstrate that serum-stimulated oligodendrocyte differentiation is enhanced by the inhibition of Notch1-associated functions.

13.
Cell Signal ; 18(4): 519-30, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16098715

ABSTRACT

Recent in vivo transplantation studies have shown that mesenchymal stem cells (MSCs) were able to differentiate into mesoderm-derived cell types as well as cells with neuroectodermal characteristics, suggesting that transdifferentiation occurs in the mammalian system. We have reported an immortalized line of human MSCs (hMSCs), KP-hMSCs, which expresses CD29, CD44, CD90, and CD105, and complies with the characteristics shared by mere hMSCs. In a current experiment, we further demonstrated that expanded KP-hMSCs exhibited markers of neuroepithelial or neural precursor cells, such as Nestin, Musashi-1, Vimentin, NCAM, Pax-6, and Sox-9. KP-hMSCs simultaneously expressed proteins of the neuronal, astrocyte, and oligodendrocyte lineages during culture expansion; in addition, they initiated neurite outgrowth and eradicated protein expressions of astrocyte and oligodendrocyte lineages in response to the elevated signaling of the cAMP-PKA pathway after serum depletion in a defined neural induction medium. From the current results, KP-hMSCs may be used to elucidate molecular signaling on the neural differentiation of adult human non-neural tissues. We also presented evidence for the possibility that adult MSCs and fetal neuroepithelial or neural precursor cells both provide for the continual maintenance and repair of the postnatal neural tissues and may derive from the same origin or have one deriving from the other.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neurites/metabolism , Signal Transduction/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Mesenchymal Stem Cells/drug effects , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/drug effects , Phosphorylation , Signal Transduction/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...