Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Int J Mol Sci ; 25(5)2024 Mar 01.
Article En | MEDLINE | ID: mdl-38474134

The slmap gene is alternatively spliced to generate many isoforms that are abundant in developing myocardium. The largest protein isoform SLMAP3 is ubiquitously expressed and has been linked to cardiomyopathy, Brugada syndrome and Hippo signaling. To examine any role in cardiogenesis, mice homozygous for floxed slmap allele were crossed with Nkx2.5-cre mice to nullify its expression in cardiac progenitors. Targeted deletion of the slmap gene resulted in the specific knockout (KO) of the SLMAP3 (~91 KDa) isoform without any changes in the expression of the SLMAP2 (~43 kDa) or the SLMAP1 (~35 kDa) isoforms which continued to accumulate to similar levels as seen in Wt embryonic hearts. The loss of SLMAP3 from cardiac progenitors resulted in decreased size of the developing embryonic hearts evident at E9.5 to E16.5 with four small chambers and significantly thinner left ventricles. The proliferative capacity assessed with the phosphorylation of histone 3 or with Ki67 in E12.5 hearts was not significantly altered due to SLMAP3 deficiency. The size of embryonic cardiomyocytes, marked with anti-Troponin C, revealed significantly smaller cells, but their hypertrophic response (AKT1 and MTOR1) was not significantly affected by the specific loss of SLMAP3 protein. Further, no changes in phosphorylation of MST1/2 or YAP were detected in SLMAP3-KO embryonic myocardium, ruling out any impact on Hippo signaling. Rat embryonic cardiomyocytes express the three SLMAP isoforms and their knockdown (KD) with sh-RNA, resulted in decreased proliferation and enhanced senescence but without any impact on Hippo signaling. Collectively, these data show that SLMAP is critical for normal cardiac development with potential for the various isoforms to serve compensatory roles. Our data imply novel mechanisms for SLMAP action in cardiac growth independent of Hippo signaling.


Hippo Signaling Pathway , Myocardium , Mice , Rats , Animals , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Membrane Proteins/metabolism , Protein Isoforms/metabolism
2.
PLoS One ; 14(4): e0214669, 2019.
Article En | MEDLINE | ID: mdl-30934005

The sarcolemmal membrane associated proteins (SLMAPs) belong to the super family of tail anchored membrane proteins which serve diverse roles in biology including cell growth, protein trafficking and ion channel regulation. Mutations in human SLMAP have been linked to Brugada syndrome with putative deficits in trafficking of the sodium channel (Nav1.5) to the cell membrane resulting in aberrant electrical activity and heart function. Three main SLMAP isoforms (SLMAP1 (35 kDa), SLMAP2 (45 kDa), and SLMAP3 (91 kDa)) are expressed in myocardium but their precise role remains to be defined. Here we generated transgenic (Tg) mice with cardiac-specific expression of the SLMAP3 isoform during postnatal development which present with a significant decrease (20%) in fractional shortening and (11%) in cardiac output at 5 weeks of age. There was a lack of any notable cardiac remodeling (hypertrophy, fibrosis or fetal gene activation) in Tg hearts but the electrocardiogram indicated a significant increase (14%) in the PR interval and a decrease (43%) in the R amplitude. Western blot analysis indicated a selective and significant decrease (55%) in protein levels of Nav1.5 while 45% drop in its transcript levels were detectable by qRT-PCR. Significant decreases in the protein and transcript levels of the calcium transport system of the sarcoplasmic reticulum (SERCA2a/PLN) were also evident in Tg hearts. These data reveal a novel role for SLMAP3 in the selective regulation of important ion transport proteins at the level of gene expression and suggest that it may be a unique target in cardiovascular function and disease.


Heart/physiology , Membrane Proteins/physiology , Myocardium/metabolism , Animals , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism
3.
Molecules ; 24(6)2019 Mar 26.
Article En | MEDLINE | ID: mdl-30917606

The genetic modification of the mouse genome using the cre-lox system has been an invaluable tool in deciphering gene and protein function in a temporal and/or spatial manner. However, it has its pitfalls, as researchers have shown that the unregulated expression of cre recombinase can cause DNA damage, the consequences of which can be very detrimental to mouse health. Previously published literature on the most utilized cardiac-specific cre, αMHC-cre, mouse model exhibited a nonlethal hypertrophic cardiomyopathy (HCM) with aging. However, using the same αMHC-cre mice, we observed a cardiac pathology, resulting in complete lethality by 11 months of age. Echocardiography and histology revealed that the αMHC-cre mice were displaying symptoms of dilated cardiomyopathy (DCM) by seven months of age, which ultimately led to their demise in the absence of any HCM at any age. Molecular analysis showed that this phenotype was associated with the DNA damage response through the downregulation of activated p38 and increased expression of JNK, p53, and Bax, known inducers of myocyte death resulting in fibrosis. Our data urges strong caution when interpreting the phenotypic impact of gene responses using αMHC-cre mice, since a lethal DCM was induced by the cre driver in an age-dependent manner in this commonly utilized model system.


Aging/genetics , Cardiomyopathy, Dilated/diagnostic imaging , Integrases/metabolism , Myosin Heavy Chains/genetics , Aging/metabolism , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , DNA Damage , Disease Models, Animal , Echocardiography , Gene Expression Regulation , Genes, Lethal , Integrases/genetics , Mice , Myosin Heavy Chains/metabolism , Phenotype
4.
Cell Signal ; 40: 230-238, 2017 12.
Article En | MEDLINE | ID: mdl-28964969

The E2F/Rb pathway regulates cell growth, differentiation, and death. In particular, E2F1 promotes apoptosis in all cells including those of the heart. E2F6, which represses E2F activity, was found to induce dilated cardiomyopathy in the absence of apoptosis in murine post-natal heart. Here we evaluate the anti-apoptotic potential of E2F6 in neonatal cardiomyocytes (NCM) from E2F6-Tg hearts which showed significantly less caspase-3 cleavage, a lower Bax/Bcl2 ratio, and improved cell viability in response to CoCl2 exposure. This correlated with a decrease in the pro-apoptotic E2F3 protein levels. In contrast, no difference in apoptotic markers or cell viability was observed in response to Doxorubicin (Dox) treatment between Wt and Tg-NCM. Dox caused a rapid and dramatic loss of the E2F6 protein in Tg-NCM within 6h and was undetectable after 12h. The level of e2f6 transcript was unchanged in Wt NCM, but was dramatically decreased in Tg cells in response to both Dox and CoCl2. This was related to an impact of the drugs on the α-myosin heavy chain promoter used to drive the E2F6 transgene. By comparison in HeLa, Dox induced apoptosis through upregulation of endogenous E2F1 involving post-transcriptional mechanisms, while E2F6 was down regulated with induction of the Checkpoint kinase-1 and proteasome degradation. These data imply that E2F6 serves to modulate E2F activity and protect cells including cardiomyocytes from apoptosis and improve survival. Strategies to modulate E2F6 levels may be therapeutically useful to mitigate cell death associated disorders.


Apoptosis/genetics , E2F1 Transcription Factor/genetics , E2F6 Transcription Factor/genetics , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Cardiomyopathy, Dilated , Caspase 3/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Doxorubicin/administration & dosage , E2F3 Transcription Factor/genetics , Gene Expression Regulation, Developmental/drug effects , HeLa Cells , Humans , Mice , Myocytes, Cardiac/drug effects
5.
PLoS One ; 12(1): e0170066, 2017.
Article En | MEDLINE | ID: mdl-28085920

RATIONALE: The E2F pathway plays a critical role in cardiac growth and development, yet its role in cardiac metabolism remains to be defined. Metabolic changes play important roles in human heart failure and studies imply the ketogenic enzyme ß-hydroxybutyrate dehydrogenase I (BDH1) is a potential biomarker. OBJECTIVE: To define the role of the E2F pathway in cardiac metabolism and dilated cardiomyopathy (DCM) with a focus on BDH1. METHODS AND RESULTS: We previously developed transgenic (Tg) mice expressing the transcriptional repressor, E2F6, to interfere with the E2F/Rb pathway in post-natal myocardium. These Tg mice present with an E2F6 dose dependent DCM and deregulated connexin-43 (CX-43) levels in myocardium. Using the Seahorse platform, a 22% decrease in glycolysis was noted in neonatal cardiomyocytes isolated from E2F6-Tg hearts. This was associated with a 39% reduction in the glucose transporter GLUT4 and 50% less activation of the regulator of glucose metabolism AKT2. The specific reduction of cyclin B1 (70%) in Tg myocardium implicates its importance in supporting glycolysis in the postnatal heart. No changes in cyclin D expression (known to regulate mitochondrial activity) were noted and lipid metabolism remained unchanged in neonatal cardiomyocytes from Tg hearts. However, E2F6 induced a 40-fold increase of the Bdh1 transcript and 890% increase in its protein levels in hearts from Tg pups implying a potential impact on ketolysis. By contrast, BDH1 expression is not activated until adulthood in normal myocardium. Neonatal cardiomyocytes from Wt hearts incubated with the ketone ß-hydroxybutyrate (ß-OHB) showed a 100% increase in CX-43 protein levels, implying a role for ketone signaling in gap junction biology. Neonatal cardiomyocyte cultures from Tg hearts exhibited enhanced levels of BDH1 and CX-43 and were not responsive to ß-OHB. CONCLUSIONS: The data reveal a novel role for the E2F pathway in regulating glycolysis in the developing myocardium through a mechanism involving cyclin B1. We reveal BDH1 expression as an early biomarker of heart failure and its potential impact, through ketone signaling, on CX-43 levels in E2F6-induced DCM.


Cardiomyopathy, Dilated/physiopathology , E2F6 Transcription Factor/metabolism , Glycolysis/physiology , Hydroxybutyrate Dehydrogenase/metabolism , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology
6.
J Mol Cell Cardiol ; 84: 179-90, 2015 Jul.
Article En | MEDLINE | ID: mdl-25944088

The E2F/Pocket protein (Rb) pathway regulates cell growth, differentiation, and death by modulating gene expression. We previously examined this pathway in the myocardium via manipulation of the unique E2F repressor, E2F6, which is believed to repress gene activity independently of Rb. Mice with targeted expression of E2F6 in postnatal myocardium developed dilated cardiomyopathy (DCM) without hypertrophic growth. We assessed the mechanisms of the apparent failure of compensatory hypertrophic growth as well as their response to the ß-adrenergic agonist isoproterenol. As early as 2 weeks, E2F6 transgenic (Tg) mice present with dilated thinner left ventricles and significantly reduced ejection fraction and fractional shortening which persists at 6 weeks of age, but with no apparent increase in left ventricle weight: body weight (LVW:BW). E2F6-Tg mice treated with isoproterenol (6.1 mg/kg/day) show double the increase in LVW:BW than their Wt counterparts (32% vs 16%, p-value: 0.007). Western blot analysis revealed the activation of the adrenergic pathway in Tg heart tissue under basal conditions with ~2-fold increase in the level of ß2-adrenergic receptors (p-value: 8.9E-05), protein kinase A catalytic subunit (PKA-C) (p-value: 0.0176), activated c-Src tyrosine-protein kinase (p-value: 0.0002), extracellular receptor kinase 2 (ERK2) (p-value: 0.0005), and induction of the anti-apoptotic protein Bcl2 (p-value 0. 0.00001). In contrast, a ~60% decrease in the cardiac growth regulator: AKT1 (p-value 0.0001) and a ~four fold increase in cyclic AMP dependent phosphodiesterase 4D (PDE4D), the negative regulator of PKA activity, were evident in the myocardium of E2F6-Tg mice. The expression of E2F3 was down-regulated by E2F6, but was restored by isoproterenol. Further, Rb expression was down-regulated in Tg mice in response to isoproterenol implying a net activation of the E2F pathway. Thus the unique regulation of E2F activity by E2F6 renders the myocardium hypersensitive to adrenergic stimulus resulting in robust hypertrophic growth. These data reveal a novel interplay between the E2F pathway, ß2-adrenergic/PKA/PDE4D, and ERK/c-Src axis in fine tuning the pathological hypertrophic growth response. E2F6 deregulates E2F3 such that pro-hypertrophic growth and survival are enhanced via ß2-adrenergic signaling however this response is outweighed by the induction of anti-hypertrophic signals so that left ventricle dilation proceeds without any increase in muscle mass.


Cardiomegaly/metabolism , Cardiomegaly/pathology , E2F6 Transcription Factor/metabolism , Myocardium/metabolism , Myocardium/pathology , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Animals , Calcium-Binding Proteins/metabolism , Cardiomegaly/complications , Cardiomegaly/enzymology , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Survival , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Down-Regulation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Isoproterenol , Mice, Transgenic , Myocardium/enzymology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Adrenergic, beta/genetics , Retinoblastoma Protein/metabolism , src-Family Kinases/metabolism
7.
FASEB J ; 26(6): 2569-79, 2012 Jun.
Article En | MEDLINE | ID: mdl-22403008

The E2F/Rb pathway regulates cardiac growth and development and holds great potential as a therapeutic target. The E2F6 repressor is a unique E2F member that acts independently of pocket proteins. Forced expression of E2F6 in mouse myocardium induced heart failure and mortality, with severity of symptoms correlating to E2F6 levels. Echocardiography demonstrated a 37% increase (P<0.05) in left ventricular end-diastolic diameter and reduced ejection fraction (<40%, P<0.05) in young transgenic (Tg) mice. Microarray and qPCR analysis revealed a paradoxical increase in E2F-responsive genes, which regulate the cell cycle, without changes in cardiomyocyte cell number or size in Tg mice. Young adult Tg mice displayed a 75% (P<0.01) decrease in gap junction protein connexin-43, resulting in abnormal electrocardiogram including a 24% (P<0.05) increase in PR interval. Further, mir-206, which targets connexin-43, was up-regulated 10-fold (P<0.05) in Tg myocardium. The mitogen-activated protein kinase pathway, which regulates the levels of miR-206 and connexin-43, was activated in Tg hearts. Thus, deregulated E2F6 levels evoked abnormal gene expression at transcriptional and post-transcriptional levels, leading to cardiac remodeling and dilated cardiomyopathy. The data highlight an unprecedented role for the strict regulation of the E2F pathway in normal postnatal cardiac function.


Cardiomyopathy, Dilated/etiology , E2F6 Transcription Factor/physiology , Animals , Connexin 43/biosynthesis , Down-Regulation , Gene Expression/drug effects , Heart Failure/physiopathology , Mice , Mice, Transgenic , Myocardium/metabolism
8.
Am J Physiol Heart Circ Physiol ; 302(5): H1138-45, 2012 Mar 01.
Article En | MEDLINE | ID: mdl-22180652

Sarcolemmal membrane-associated proteins (SLMAPs) are components of cardiac membranes involved in excitation-contraction (E-C) coupling. Here, we assessed the role of SLMAP in cardiac structure and function. We generated transgenic (Tg) mice with cardiac-restricted overexpression of SLMAP1 bearing the transmembrane domain 2 (TM2) to potentially interfere with endogenous SLMAP through homodimerization and subcellular targeting. Histological examination revealed vacuolated myocardium; the severity of which correlated with the expression level of SLMAP1-TM2. High resolution microscopy showed dilation of the sarcoplasmic reticulum/endoplasmic reticulum (SR/ER) and confocal imaging combined with biochemical analysis indicated targeting of SLMAP1-TM2 to the SR/ER membranes and inappropriate homodimerization. Older (28 wk of age) Tg mice exhibited reduced contractility with impaired relaxation as assessed by left ventricle pressure monitoring. The ventricular dysfunction was associated with electrophysiological abnormalities (elongated QT interval). Younger (5 wk of age) Tg mice also exhibited an elongated QT interval with minimal functional disturbances associated with the activation of the fetal gene program. They were less responsive to isoproterenol challenge (ΔdP/dt(max)) and developed electrical and left ventricular pressure alternans. The altered electrophysiological and functional disturbances in Tg mice were associated with diminished expression level of calcium cycling proteins of the sarcoplasmic reticulum such as the ryanodine receptor, Ca(2+)-ATPase, calsequestrin, and triadin (but not phospholamban), as well as significantly reduced calcium uptake in microsomal fractions. These data demonstrate that SLMAP is a regulator of E-C coupling at the level of the SR and its perturbation results in progressive deterioration of cardiac electrophysiology and function.


Heart/physiology , Membrane Proteins/physiology , Sarcoplasmic Reticulum/physiology , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/metabolism , Calcium-Transporting ATPases/biosynthesis , Calsequestrin/biosynthesis , Carrier Proteins/biosynthesis , Female , Isoproterenol/pharmacology , Membrane Proteins/genetics , Mice , Mice, Transgenic , Muscle Proteins/biosynthesis , Myocardial Contraction/physiology , Myocardium/cytology , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/biosynthesis , Sarcoplasmic Reticulum/metabolism
9.
J Biol Chem ; 284(41): 28212-28221, 2009 Oct 09.
Article En | MEDLINE | ID: mdl-19671701

The sarco-endoplasmic reticulum calcium ATPase 2a (SERCA2a) is critical for sequestering cytosolic calcium into the sarco-endoplasmic reticulum (SR) and regulating cardiac muscle relaxation. Protein-protein interactions indicated that it exists in complex with Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and its anchoring protein alphaKAP. Confocal imaging of isolated cardiomyocytes revealed the colocalization of CAMKII and alphaKAP with SERCA2a at the SR. Deletion analysis indicated that SERCA2a and CaMKII bind to different regions in the association domain of alphaKAP but not with each other. Although deletion of the putative N-terminal hydrophobic amino acid stretch in alphaKAP prevented its membrane targeting, it did not influence binding to SERCA2a or CaMKII. Both CaMKIIdelta(C) and the novel CaMKIIbeta(4) isoforms were found to exist in complex with alphaKAP and SERCA2a at the SR and were able to phosphorylate Thr-17 on phospholamban (PLN), an accessory subunit and known regulator of SERCA2a activity. Interestingly, the presence of alphaKAP was also found to significantly modulate the Ca(2+)/calmodulin-dependent phosphorylation of Thr-17 on PLN. These data demonstrate that alphaKAP exhibits a novel interaction with SERCA2a and may serve to spatially position CaMKII isoforms at the SR and to uniquely modulate the phosphorylation of PLN.


A Kinase Anchor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , A Kinase Anchor Proteins/genetics , Animals , Calcium-Binding Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calmodulin/metabolism , HeLa Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Microsomes/enzymology , Myocardium/cytology , Myocardium/enzymology , Phosphorylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sequence Deletion
10.
BMC Cell Biol ; 10: 48, 2009 Jun 19.
Article En | MEDLINE | ID: mdl-19538755

BACKGROUND: Tail anchored (TA) membrane proteins target subcellular structures via a C-terminal transmembrane domain and serve prominent roles in membrane fusion and vesicle transport. Sarcolemmal Membrane Associated Protein (SLMAP) possesses two alternatively spliced tail anchors (TA1 or TA2) but their specificity of subcellular targeting remains unknown. RESULTS: TA1 or TA2 can direct SLMAP to reticular structures including the endoplasmic reticulum (ER), whilst TA2 directs SLMAP additionally to the mitochondria. Despite the general structural similarity of SLMAP to other vesicle trafficking proteins, we found no evidence for its localization with the vesicle transport machinery or a role in vesicle transport. The predicted transmembrane region of TA2 is flanked on either side by a positively charged amino acid and is itself less hydrophobic than the transmembrane helix present in TA1. Substitution of the positively charged amino acids, in the regions flanking the transmembrane helix of TA2, with leucine did not alter its subcellular targeting. The targeting of SLMAP to the mitochondria was dependent on the hydrophobic nature of TA2 since targeting of SLMAP-TA2 was prevented by the substitution of leucine (L) for moderately hydrophobic amino acid residues within the transmembrane region. The SLMAP-TA2-4L mutant had a hydrophobic profile that was comparable to that of SLMAP-TA1 and had identical targeting properties to SLMAP-TA1. CONCLUSION: Thus the overall hydrophobicity of the two alternatively spliced TAs in SLMAP determines its subcellular targeting and TA2 predominantly directs SLMAP to the mitochondira where it may serve roles in the function of this organelle.


Membrane Proteins/chemistry , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cytoskeleton/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Hydrophobic and Hydrophilic Interactions , Intracellular Membranes/metabolism , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Protein Sorting Signals , Rats
11.
Am J Physiol Cell Physiol ; 290(1): C189-99, 2006 Jan.
Article En | MEDLINE | ID: mdl-16107498

E2F6 is believed to repress E2F-responsive genes and therefore serve a role in cell cycle regulation. Analysis of the human E2F6 promoter region revealed the presence of two putative E2F binding sites, both of which were found to be functionally critical because deletion or mutations of these sites abolished promoter activity. Ectopic expression of E2F1 protein was found to increase E2F6 mRNA levels and significantly upregulate E2F6 promoter activity. Deletion or mutation of the putative E2F binding sites nullified the effects of E2F1 on the E2F6 promoter activity. Studies on the temporal induction of E2F family members demonstrated that the activating E2Fs, and most notably E2F1, were upregulated before E2F6 during cell cycle progression at the G1/S phase, and this coincided with the time course of induction experienced by the E2F6 promoter during the course of the cell cycle. EMSAs indicated the specific binding of nuclear complexes to the E2F6 promoter that contained E2F1-related species whose binding was specifically competed by the consensus E2F binding site. Chromatin immunoprecipitation assays with anti-E2Fs demonstrated the association of E2F family members with the E2F6 promoter in vivo. These data indicate that the expression of the E2F6 repressor is influenced at the transcriptional level by E2F family members and suggest that interplay among these transcriptional regulators, especially E2F1, may be critical for cell cycle regulation.


E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F6 Transcription Factor/genetics , E2F6 Transcription Factor/metabolism , Transcriptional Activation/physiology , Base Sequence , Binding Sites/genetics , Cell Cycle/genetics , Cell Line , Gene Expression Regulation/physiology , Humans , Kidney/cytology , Molecular Sequence Data , Promoter Regions, Genetic/physiology , RNA, Messenger/analysis
12.
Mol Cell Biochem ; 270(1-2): 215-21, 2005 Feb.
Article En | MEDLINE | ID: mdl-15792370

We report the isolation of a full length cDNA from cardiac muscle that encodes a approximately 73 kDa calcium/calmodulin (CaM) dependent kinase IIbeta isoform (CaMKIIbeta(C)) that was generated by alternative splicing of the CaMKIIbeta gene. Antipeptide antibodies raised to specific regions of the kinase identified a 73 kDa kinase polypeptide in cardiac SR. Anti-alpha kinase anchoring protein (alphaKAP) antibodies identified a 25 kDa polypeptide in cardiac SR and RT-PCR followed by sequence analysis confirmed the presence of a full length alphaKAP encoding transcript in myocardium. Protein interaction assays revealed that the 73 kDa CaMKIIbeta(C) binds GAPDH to modulate the production of NADH in a Ca2+/CaM dependent reaction. The presence of a CaMKIIbeta isoform that can target the SR presumably via its membrane anchor alphaKAP defines a previously unrecognized Ca2+/CaM regulatory system in myocardium.


Alternative Splicing , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Myocardium/metabolism , Myocardium/pathology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cells, Cultured , Cytosol/metabolism , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Exons , Gene Library , Glutathione Transferase/metabolism , Humans , Immunoblotting , Introns , Molecular Sequence Data , Muscle, Skeletal/metabolism , Myocardium/enzymology , NAD/metabolism , Peptides/chemistry , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Rabbits , Sarcoplasmic Reticulum/metabolism , Sequence Homology, Amino Acid , Tissue Distribution
13.
Am J Physiol Heart Circ Physiol ; 288(4): H1810-9, 2005 Apr.
Article En | MEDLINE | ID: mdl-15591093

The spatial arrangement of the cell-surface membranes (sarcolemma and transverse tubules) and internal membranes of the sarcoplasmic reticulum relative to the myofibril is critical for effective excitation-contraction (E-C) coupling in cardiac myocytes; however, the molecular determinants of this order remain to be defined. Here, we ascribe molecular and cellular properties to the coiled-coil, tail-anchored sarcolemmal membrane-associated protein (SLMAP) that are consistent with a potential role in organizing the E-C coupling apparatus of the cardiomyocyte. The expression of SLMAP was developmentally regulated and its localization was distinctly apparent at the level of the membranes involved in regulating the E-C coupling mechanism. Several SLMAP isoforms were expressed in the cardiac myocyte with unique COOH-terminal membrane anchors that could target this molecule to distinct subcellular membranes. Protein interaction analysis indicated that SLMAPs could self assemble and bind myosin in cardiac muscle. The cardiac-specific expression of SLMAP isoforms that can be targeted to distinct subcellular membranes, self assemble, and interact with the myofibril suggests a potential role for this molecule in the structural arrangement of the E-C coupling apparatus.


Membrane Proteins/chemistry , Membrane Proteins/genetics , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum/physiology , Age Factors , Animals , COS Cells , Chlorocebus aethiops , Female , Gene Expression Regulation, Developmental , Heart/growth & development , Heart/physiology , Membrane Proteins/metabolism , Mice , Pregnancy , Protein Structure, Tertiary , Sarcolemma/physiology , Structure-Activity Relationship , Transfection
14.
J Biol Chem ; 279(34): 35176-82, 2004 Aug 20.
Article En | MEDLINE | ID: mdl-15199064

The skeletal muscle specific Ca(2)+/calmodulin-dependent protein kinase (CaMKIIbeta(M)) is localized to the sarcoplasmic reticulum (SR) by an anchoring protein, alphaKAP, but its function remains to be defined. Protein interactions of CaMKIIbeta(M) indicated that it exists in complex with enzymes involved in glycolysis at the SR membrane. The kinase was found to complex with glycogen phosphorylase, glycogen debranching enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and creatine kinase in the SR membrane. CaMKIIbeta(M) was also found to assemble with aldolase A, GAPDH, enolase, lactate dehydrogenase, creatine kinase, pyruvate kinase, and phosphorylase b kinase from the cytosolic fraction. The interacting proteins were substrates of CaMKIIbeta(M), and their phosphorylation was enhanced in a Ca(2+)- and calmodulin (CaM)-dependent manner. The CaMKIIbeta(M) could directly phosphorylate GAPDH and markedly increase ( approximately 3.4-fold) its activity in a Ca(2+)/CaM-dependent manner. These data suggest that the muscle CaMKIIbeta(M) isoform may serve to assemble the glycogen-mobilizing and glycolytic enzymes at the SR membrane and specifically modulate the activity of GAPDH in response to calcium signaling. Thus, the activation of CaMKIIbeta(M) in response to calcium signaling would serve to modulate GAPDH and thereby ATP and NADH levels at the SR membrane, which in turn will regulate calcium transport processes.


Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Muscle, Skeletal/enzymology , Sarcoplasmic Reticulum/enzymology , Animals , Calcium Signaling , Enzyme Activation , Isoenzymes/metabolism , Models, Molecular , Muscle, Skeletal/ultrastructure , Phosphorylation , Rabbits , Rats
15.
J Cell Sci ; 117(Pt 11): 2271-81, 2004 May 01.
Article En | MEDLINE | ID: mdl-15126628

The microtubule organizing centre (MTOC) or the centrosome serves a crucial role in the establishment of cellular polarity, organization of interphase microtubules and the formation of the bipolar mitotic spindle. We have elucidated the genomic structure of a gene encoding the sarcolemmal membrane-associated protein (SLMAP), which encodes a 91 kDa polypeptide with a previously uncharacterized N-terminal sequence encompassing a forkhead-associated (FHA) domain that resides at the centrosome. Anti-peptide antibodies directed against SLMAP N-terminal sequences showed colocalization with gamma-tubulin at the centrosomes at all phases of the cell cycle. Agents that specifically disrupt microtubules did not affect SLMAP association with centrosomes. Furthermore, SLMAP sequences directed a reporter green fluorescent protein (GFP) to the centrosome, and deletions of the newly identified N-terminal sequence from SLMAP prevented the centrosomal targeting. Deletion-mutant analysis concluded that overall, structural determinants in SLMAP were responsible for centrosomal targeting. Elevated levels of centrosomal SLMAP were found to be lethal, whereas mutants that lacked centrosomal targeting inhibited cell growth accompanied by an accumulation of cells at the G2/M phase of the cell cycle.


Centrosome/chemistry , Centrosome/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Cell Proliferation , Cloning, Molecular , Cytoplasm/metabolism , Genomic Library , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Nuclear Envelope/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Transfection
16.
Biochem J ; 381(Pt 3): 599-608, 2004 Aug 01.
Article En | MEDLINE | ID: mdl-15086317

Sarcolemmal-membrane-associated proteins (SLMAPs) define a new class of coiled-coil tail-anchored membrane proteins generated by alternative splicing mechanisms. An in vivo expression analysis indicated that SLMAPs are present in somites (11 days post-coitum) as well as in fusing myotubes and reside at the level of the sarcoplasmic reticulum and transverse tubules in adult skeletal muscles. Skeletal-muscle myoblasts were found to express a single 5.9 kb transcript, which encodes the full-length approximately 91 kDa SLMAP3 isoform. Myoblast differentiation was accompanied by the stable expression of the approximately 91 kDa SLMAP protein as well as the appearance of an approximately 80 kDa isoform. Deregulation of SLMAPs by ectopic expression in myoblasts resulted in a potent inhibition of fusion without affecting the expression of muscle-specific genes. Membrane targeting of the de-regulated SLMAPs was not critical for the inhibition of myotube development. Protein-protein interaction assays indicated that SLMAPs are capable of self-assembling, and the de-regulated expression of mutants that were not capable of forming SLMAP homodimers also inhibited myotube formation. These results imply that regulated levels and the temporal induction of SLMAP isoforms are important for normal muscle development.


Gene Expression Regulation, Developmental/physiology , Membrane Proteins/genetics , Muscle, Skeletal/embryology , Myoblasts, Skeletal/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Dimerization , Embryo, Mammalian/chemistry , Female , Leucine-Rich Repeat Proteins , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Molecular Weight , Myoblasts, Skeletal/chemistry , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Pregnancy , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Proteins/metabolism , Time Factors
...