Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Channels (Austin) ; 11(6): 604-615, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28901828

ABSTRACT

L-type-voltage-dependent Ca2+ channels (L-VDCCs; CaV1.2, α1C), crucial in cardiovascular physiology and pathology, are modulated via activation of G-protein-coupled receptors and subsequently protein kinase C (PKC). Despite extensive study, key aspects of the mechanisms leading to PKC-induced Ca2+ current increase are unresolved. A notable residue, Ser1928, located in the distal C-terminus (dCT) of α1C was shown to be phosphorylated by PKC. CaV1.2 undergoes posttranslational modifications yielding full-length and proteolytically cleaved CT-truncated forms. We have previously shown that, in Xenopus oocytes, activation of PKC enhances α1C macroscopic currents. This increase depended on the isoform of α1C expressed. Only isoforms containing the cardiac, long N-terminus (L-NT), were upregulated by PKC. Ser1928 was also crucial for the full effect of PKC. Here we report that, in Xenopus oocytes, following PKC activation the amount of α1C protein expressed in the plasma membrane (PM) increases within minutes. The increase in PM content is greater with full-length α1C than in dCT-truncated α1C, and requires Ser1928. The same was observed in HL-1 cells, a mouse atrium cell line natively expressing cardiac α1C, which undergoes the proteolytic cleavage of the dCT, thus providing a native setting for exploring the effects of PKC in cardiomyocytes. Interestingly, activation of PKC preferentially increased the PM levels of full-length, L-NT α1C. Our findings suggest that part of PKC regulation of CaV1.2 in the heart involves changes in channel's cellular fate. The mechanism of this PKC regulation appears to involve the C-terminus of α1C, possibly corroborating the previously proposed role of NT-CT interactions within α1C.


Subject(s)
Calcium Channels, L-Type/biosynthesis , Cell Membrane/metabolism , Protein Kinase C/metabolism , Animals , Cells, Cultured , Mice , Xenopus laevis
2.
J Vasc Res ; 54(3): 131-142, 2017.
Article in English | MEDLINE | ID: mdl-28468000

ABSTRACT

AIM: Vascular remodeling following injury substantially accounts for restenosis and adverse clinical outcomes. In this study, we investigated the role of the giant scaffold protein Ahnak1 in vascular healing after endothelial denudation of the murine femoral artery. METHODS: The spatiotemporal expression pattern of Ahnak1 and Ahnak2 was examined using specific antibodies and real-time quantitative PCR. Following wire-mediated endothelial injury of Ahnak1-deficient mice and wild-type (WT) littermates, the processes of vascular healing were analyzed. RESULTS: Ahnak1 and Ahnak2 showed a mutually exclusive vascular expression pattern, with Ahnak1 being expressed in the endothelium and Ahnak2 in the medial cells in naïve WT arteries. After injury, a marked increase of Ahnak1- and Ahnak2-positive cells at the lesion site became evident. Both proteins showed a strong upregulation in neointimal cells 14 days after injury. Ahnak1-deficient mice showed delayed vascular healing and dramatically impaired re-endothelialization that resulted in prolonged adverse vascular remodeling, when compared to the WT littermates. CONCLUSION: The large scaffold and adaptor proteins Ahnak1 and Ahnak2 exhibit differential expression patterns and functions in naïve and injured arteries. Ahnak1 plays a nonredundant protective role in vascular healing.


Subject(s)
Femoral Artery/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Vascular Remodeling , Vascular System Injuries/metabolism , Wound Healing , Animals , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Femoral Artery/injuries , Femoral Artery/pathology , Genotype , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phenotype , Re-Epithelialization , Time Factors , Vascular System Injuries/genetics , Vascular System Injuries/pathology
3.
J Physiol ; 595(10): 3181-3202, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28194788

ABSTRACT

KEY POINTS: ß-Adrenergic stimulation enhances Ca2+ entry via L-type CaV 1.2 channels, causing stronger contraction of cardiac muscle cells. The signalling pathway involves activation of protein kinase A (PKA), but the molecular details of PKA regulation of CaV 1.2 remain controversial despite extensive research. We show that PKA regulation of CaV 1.2 can be reconstituted in Xenopus oocytes when the distal C-terminus (dCT) of the main subunit, α1C , is truncated. The PKA upregulation of CaV 1.2 does not require key factors previously implicated in this mechanism: the clipped dCT, the A kinase-anchoring protein 15 (AKAP15), the phosphorylation sites S1700, T1704 and S1928, or the ß subunit of CaV 1.2. The gating element within the initial segment of the N-terminus of the cardiac isoform of α1C is essential for the PKA effect. We propose that the regulation described here is one of two or several mechanisms that jointly mediate the PKA regulation of CaV 1.2 in the heart. ABSTRACT: ß-Adrenergic stimulation enhances Ca2+ currents via L-type, voltage-gated CaV 1.2 channels, strengthening cardiac contraction. The signalling via ß-adrenergic receptors (ß-ARs) involves elevation of cyclic AMP (cAMP) levels and activation of protein kinase A (PKA). However, how PKA affects the channel remains controversial. Recent studies in heterologous systems and genetically engineered mice stress the importance of the post-translational proteolytic truncation of the distal C-terminus (dCT) of the main (α1C ) subunit. Here, we successfully reconstituted the cAMP/PKA regulation of the dCT-truncated CaV 1.2 in Xenopus oocytes, which previously failed with the non-truncated α1C . cAMP and the purified catalytic subunit of PKA, PKA-CS, injected into intact oocytes, enhanced CaV 1.2 currents by ∼40% (rabbit α1C ) to ∼130% (mouse α1C ). PKA blockers were used to confirm specificity and the need for dissociation of the PKA holoenzyme. The regulation persisted in the absence of the clipped dCT (as a separate protein), the A kinase-anchoring protein AKAP15, and the phosphorylation sites S1700 and T1704, previously proposed as essential for the PKA effect. The CaV ß2b subunit was not involved, as suggested by extensive mutagenesis. Using deletion/chimeric mutagenesis, we have identified the initial segment of the cardiac long-N-terminal isoform of α1C as a previously unrecognized essential element involved in PKA regulation. We propose that the observed regulation, that exclusively involves the α1C subunit, is one of several mechanisms underlying the overall PKA action on CaV 1.2 in the heart. We hypothesize that PKA is acting on CaV 1.2, in part, by affecting a structural 'scaffold' comprising the interacting cytosolic N- and C-termini of α1C .


Subject(s)
Calcium Channels, L-Type/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Oocytes/physiology , Protein Subunits/physiology , Animals , Cyclic AMP/physiology , Xenopus laevis
4.
Biochem Biophys Res Commun ; 450(1): 464-9, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24911555

ABSTRACT

The essential myosin light chain (ELC) is involved in modulation of force generation of myosin motors and cardiac contraction, while its mechanism of action remains elusive. We hypothesized that ELC could modulate myosin stiffness which subsequently determines its force production and cardiac contraction. Therefore, we generated heterologous transgenic mouse (TgM) strains with cardiomyocyte-specific expression of ELC with human ventricular ELC (hVLC-1; TgM(hVLC-1)) or E56G-mutated hVLC-1 (hVLC-1(E56G); TgM(E56G)). hVLC-1 or hVLC-1(E56G) expression in TgM was around 39% and 41%, respectively of total VLC-1. Laser trap and in vitro motility assays showed that stiffness and actin sliding velocity of myosin with hVLC-1 prepared from TgM(hVLC-1) (1.67 pN/nm and 2.3 µm/s, respectively) were significantly higher than myosin with hVLC-1(E56G) prepared from TgM(E56G) (1.25 pN/nm and 1.7 µm/s, respectively) or myosin with mouse VLC-1 (mVLC-1) prepared from C57/BL6 (1.41 pN/nm and 1.5 µm/s, respectively). Maximal left ventricular pressure development of isolated perfused hearts in vitro prepared from TgM(hVLC-1) (80.0 mmHg) were significantly higher than hearts from TgM(E56G) (66.2 mmHg) or C57/BL6 (59.3±3.9 mmHg). These findings show that ELCs decreased myosin stiffness, in vitro motility, and thereby cardiac functions in the order hVLC-1>hVLC-1(E56G)≈mVLC-1. They also suggest a molecular pathomechanism of hypertrophic cardiomyopathy caused by hVLC-1 mutations.


Subject(s)
Heart/physiology , Myocardial Contraction/physiology , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Animals , Elastic Modulus , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/physiology , Molecular Motor Proteins/ultrastructure , Myosin Light Chains/ultrastructure , Structure-Activity Relationship , Tensile Strength/physiology
5.
Cardiovasc Res ; 101(3): 533-42, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24336332

ABSTRACT

AIM: Nitric oxide (NO) synthesized by cardiomyocytes plays an important role in the regulation of cardiac function. Here, we studied the impact of NO signalling on calcium influx in human right atrial myocytes and its relation to atrial fibrillation (AF). METHODS AND RESULTS: Right atrial appendages (RAAs) were obtained from patients in sinus rhythm (SR) and AF. The biotin-switch technique was used to evaluate endogenous S-nitrosylation of the α1C subunit of L-type calcium channels. Comparing SR to AF, S-nitrosylation of Ca(2+) channels was similar. Direct effects of the NO donor S-nitroso-N-acetyl-penicillamine (SNAP) on L-type calcium current (ICa,L) were studied in cardiomyocytes with standard voltage-clamp techniques. In SR, ICa,L increased with SNAP (100 µM) by 48%, n/N = 117/56, P < 0.001. The SNAP effect on ICa,L involved activation of soluble guanylate cyclase and protein kinase A. Specific inhibition of phosphodiesterase (PDE)3 with cilostamide (1 µM) enhanced ICa,L to a similar extent as SNAP. However, when cAMP was elevated by PDE3 inhibition or ß-adrenoceptor stimulation, SNAP reduced ICa,L, pointing to cGMP-cAMP cross-regulation. In AF, the stimulatory effect of SNAP on ICa,L was attenuated, while its inhibitory effect on isoprenaline- or cilostamide-stimulated current was preserved. cGMP elevation with SNAP was comparable between the SR and AF group. Moreover, the expression of PDE3 and soluble guanylate cyclase was not reduced in AF. CONCLUSION: NO exerts dual effects on ICa,L in SR with an increase of basal and inhibition of cAMP-stimulated current, and in AF NO inhibits only stimulated ICa,L. We conclude that in AF, cGMP regulation of PDE2 is preserved, but regulation of PDE3 is lost.


Subject(s)
Atrial Fibrillation/metabolism , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Nitric Oxide/metabolism , Atrial Fibrillation/drug therapy , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Humans , Myocardium/metabolism , Patch-Clamp Techniques/methods , Phosphodiesterase 3 Inhibitors/pharmacology , Quinolones/pharmacology
6.
Hypertension ; 62(5): 966-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24001898

ABSTRACT

Alzheimer disease features amyloid-ß (Aß) peptide deposition in brain and blood vessels and is associated with hypertension. Aß peptide can cause vasoconstriction and endothelial dysfunction. We observed that Aß peptides exert a chronotropic effect in neonatal cardiomyocytes, similar to α1-adrenergic receptor autoantibodies that we described earlier. Recently, it was shown that α1-adrenergic receptor could impair blood-brain flow. We hypothesized that Aß peptides might elicit a signal transduction pathway in vascular cells, induced by α1-adrenergic receptor activation. Aß (25-35) and Aß (10-35) induced a positive chronotropic effect in the cardiac contraction assay (28.75±1.15 and 29.40±0.98 bpm), which was attenuated by α1-adrenergic receptor blockers (urapidil, 1.53±1.17 bpm; prazosin, 0.30±0.96 bpm). Both Aß peptides induced an intracellular calcium release in vascular smooth muscle cells. Chronotropic activity and calcium response elicited by Aß (25-35) were blocked with peptides corresponding to the first extracellular loop of the α1-adrenergic receptor. We observed an induction of extracellular-regulated kinase 1/2 phosphorylation by Aß (25-35) in Chinese hamster ovary cells overexpressing α1-adrenergic receptor, vascular smooth muscle cells, and cardiomyocytes. We generated an activation-state-sensitive α1-adrenergic receptor antibody and visualized activation of the α1-adrenergic receptor by Aß peptide. Aß (25-35) induced vasoconstriction of mouse aortic rings and in coronary arteries in Langendorff-perfused rat hearts that resulted in decreased coronary flow. Both effects could be reversed by α1-adrenergic receptor blockade. Our data are relevant to the association between Alzheimer disease and hypertension. They may explain impairment of vascular responses by Aß and could have therapeutic implications.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/pharmacology , Amyloid beta-Peptides/pharmacology , Myocytes, Cardiac/drug effects , Peptide Fragments/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Heart/drug effects , Male , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
7.
J Proteomics ; 78: 72-82, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23182705

ABSTRACT

Formin-like 1 (FMNL1) is a formin-related protein highly expressed in hematopoietic cells and overexpressed in leukemias as well as diverse transformed cell lines. It has been described to play a role in diverse functions of hematopoietic cells such as phagocytosis of macrophages as well as polarization and cytotoxicity of T cells. However, the specific role of FMNL1 in these processes has not been clarified yet and regulation by interaction partners in primary hematopoietic cells has never been investigated. We performed a proteomic screen for investigation of the interactome of FMNL1 in primary hematopoietic cells resulting in the identification of a number of interaction partners. Bioinformatic analysis considering semantic similarity suggested the giant protein AHNAK1 to be an essential interaction partner of FMNL1. We confirmed AHNAK1 as a general binding partner for FMNL1 in diverse hematopoietic cells and demonstrate that the N-terminal part of FMNL1 binds to the C-terminus of AHNAK1. Moreover, we show that the constitutively activated form of FMNL1 (FMNL1γ) induces localization of AHNAK1 to the cell membrane. Finally, we provide evidence that overexpression or knock down of FMNL1 has an impact on the capacitative calcium influx after ionomycin-mediated activation of diverse cell lines and primary cells.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Cell Membrane/pathology , Female , Formins , Hematopoietic Stem Cells/pathology , Humans , K562 Cells , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Protein Transport , Proteomics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
8.
Channels (Austin) ; 6(6): 426-37, 2012.
Article in English | MEDLINE | ID: mdl-22990911

ABSTRACT

L-type voltage dependent Ca(2+) channels (L-VDCCs; Ca(v)1.2) are crucial in cardiovascular physiology. In heart and smooth muscle, hormones and transmitters operating via G(q) enhance L-VDCC currents via essential protein kinase C (PKC) involvement. Heterologous reconstitution studies in Xenopus oocytes suggested that PKC and G(q)-coupled receptors increased L-VDCC currents only in cardiac long N-terminus (NT) isoforms of α(1C), whereas known smooth muscle short-NT isoforms were inhibited by PKC and G(q) activators. We report a novel regulation of the long-NT α(1C) isoform by Gßγ. Gßγ inhibited whereas a Gßγ scavenger protein augmented the G(q)--but not phorbol ester-mediated enhancement of channel activity, suggesting that Gßγ acts upstream from PKC. In vitro binding experiments reveal binding of both Gßγ and PKC to α(1C)-NT. However, PKC modulation was not altered by mutations of multiple potential phosphorylation sites in the NT, and was attenuated by a mutation of C-terminally located serine S1928. The insertion of exon 9a in intracellular loop 1 rendered the short-NT α(1C) sensitive to PKC stimulation and to Gßγ scavenging. Our results suggest a complex antagonistic interplay between G(q)-activated PKC and Gßγ in regulation of L-VDCC, in which multiple cytosolic segments of α(1C) are involved.


Subject(s)
Calcium Channels, L-Type/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Oocytes/metabolism , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/metabolism , Xenopus laevis/metabolism , Animals , Binding Sites , Calcium Channels, L-Type/chemistry , Chloride Channels/metabolism , Enzyme Activation/drug effects , Exons/genetics , Humans , Ion Channel Gating/drug effects , Mutation/genetics , Oocytes/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Isoforms/chemistry , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Rats , Receptors, Muscarinic/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation/drug effects
9.
Biochem Biophys Res Commun ; 421(2): 184-9, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22497893

ABSTRACT

Ahnak1 has been implicated in protein kinase A (PKA)-mediated control of cardiac L-type Ca(2+) channels (Cav1.2) through its interaction with the Cavß(2) regulatory channel subunit. Here we corroborate this functional linkage by immunocytochemistry on isolated cardiomyocytes showing co-localization of ahnak1 and Cavß(2) in the T-tubule system. In previous studies Cavß(2) attachment sites which impacted the channel's PKA regulation have been located to ahnak1's proximal C-terminus (ahnak1(4889-5535), ahnak1(5462-5535)). In this study, we mapped the ahnak1-interacting regions in Cavß(2) and investigated whether Cavß(2) phosphorylation affects its binding behavior. In vitro binding assays with Cavß(2) truncation mutants and ahnak1(4889-5535) revealed that the core region of Cavß(2) consisting of Src-homology 3 (SH3), HOOK, and guanylate kinase (GK) domains was important for ahnak1 interaction while the C- and N-terminal regions were dispensable. Furthermore, Ser-296 in the GK domain of Cavß(2) was identified as novel PKA phosphorylation site by mass spectrometry. Surface plasmon resonance (SPR) binding analysis showed that Ser-296 phosphorylation did not affect the high affinity interaction (K(D)≈35 nM) between Cavß(2) and the α(1C) I-II linker, but affected ahnak1 interaction in a complex manner. SPR experiments with ahnak1(5462-5535) revealed that PKA phosphorylation of Cavß(2) significantly increased the binding affinity and, in parallel, it reduced the binding capacity. Intriguingly, the phosphorylation mimic substitution Glu-296 fully reproduced both effects, increased the affinity by ≈2.4-fold and reduced the capacity by ≈60%. Our results are indicative for the release of a population of low affinity interaction sites following Cavß(2) phosphorylation on Ser-296. We propose that this phosphorylation event is one mechanism underlying ahnak1's modulator function on Cav1.2 channel activity.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Neoplasm Proteins/metabolism , Serine/metabolism , Animals , Calcium Channels, L-Type/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Mice , Mice, Inbred Strains , Phosphorylation , Protein Interaction Maps , Protein Structure, Tertiary , Serine/genetics
10.
J Cell Biochem ; 113(3): 934-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22371973

ABSTRACT

Zonula occludens protein 1 (ZO-1) is a ubiquitous scaffolding protein, but it is unknown why it functions in very different cellular contacts. We hypothesized that a specific segment, the unique hinge region, can be bound by very different regulatory proteins. Using surface plasmon resonance spectroscopy and binding assays to peptide libraries, we show, for the first time, that the hinge region directly interacts with disparate signal elements such as G-proteins alpha 12 and alpha i2, the regulator of G-protein signaling 5, multifunctional signaling protein ahnak1, and L-type Ca2+-channel beta-2-subunit. The novel binding proteins specifically bound to a coiled coil-helix predicted in the hinge region of ZO-. The interactions were modulated by phosphorylation in the hinge helix. Activation of the G-proteins influenced their association to ZO-1. In colon cells, G alpha i2 and ZO-1 were associated, as shown by coimmunoprecipitation. After cotransfection in kidney cells, G alpha i2 barely colocalized with ZO-1; the colocalization coefficient was significantly increased when epinephrine activated G-protein signaling. In conclusion, proteins with different regulatory potential adhere to and influence cellular functions of ZO-proteins, and the interactions can be modulated via its hinge region and/or the binding proteins.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Caco-2 Cells , Cell Membrane/chemistry , Epithelial Cells/chemistry , Epithelial Cells/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , HEK293 Cells , Humans , Membrane Proteins/analysis , Membrane Proteins/chemistry , Phosphoproteins/analysis , Phosphoproteins/chemistry , RGS Proteins/metabolism , Zonula Occludens-1 Protein
11.
Cardiovasc Res ; 93(1): 200-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22038741

ABSTRACT

AIMS: The lipid and protein kinase phosphoinositide 3-kinase γ (PI3Kγ) is abundantly expressed in inflammatory cells and in the cardiovascular tissue. In recent years, its role in inflammation and in cardiac function and remodelling has been unravelled, highlighting the beneficial effects of its pharmacological inhibition. Furthermore, a role for PI3Kγ in the regulation of vascular tone has been emphasized. However, the impact of this signalling in the control of blood pressure is still poorly understood. Our study investigated the effect of a selective inhibition of PI3Kγ, obtained by using two independent small molecules, on blood pressure. Moreover, we dissected the molecular mechanisms involved in control of contraction of resistance arteries by PI3Kγ. METHODS AND RESULTS: We showed that inhibition of PI3Kγ reduced blood pressure in normotensive and hypertensive mice in a concentration-dependent fashion. This effect was dependent on enhanced vasodilatation, documented in vivo by decreased peripheral vascular resistance, and ex vivo by vasorelaxing effects on isolated resistance vessels. The vasorelaxation induced by PI3Kγ inhibition relied on blunted pressure-induced Akt phosphorylation and a myogenic contractile response. Molecular insights revealed that PI3Kγ inhibition affected smooth muscle L-type calcium channel current density and calcium influx by impairing plasma membrane translocation of the α1C L-type calcium channel subunit responsible for channel open-state probability. CONCLUSION: Overall our findings suggest that PI3Kγ inhibition could be a novel tool to modulate calcium influx in vascular smooth muscle cells, thus relaxing resistance arteries and lowering blood pressure.


Subject(s)
Blood Pressure/physiology , Calcium Channels, L-Type/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/physiology , Animals , Blood Pressure/drug effects , Calcium Signaling/drug effects , Class Ib Phosphatidylinositol 3-Kinase/genetics , Enzyme Inhibitors/pharmacology , Hypertension/drug therapy , Hypertension/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Quinoxalines/pharmacology , Thiazolidinediones/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
12.
Cardiovasc Res ; 93(3): 390-6, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22131351

ABSTRACT

AIMS: We tested the hypothesis that mutations in the human ventricular essential myosin light chain (hVLC-1) that are associated with hypertrophic cardiomyopathy (HCM) affect protein structure, binding to the IQ1 motif of cardiac myosin heavy chain (MYH) and sarcomeric sorting in neonatal cardiomyocytes. METHODS AND RESULTS: We employed circular dichroism and surface plasmon resonance spectroscopy to investigate structural properties and protein-protein interactions of a recombinant head-rod fragment of rat cardiac ß-MYH (amino acids 664-915) with alanine-mutated IQ2 domain (rß-MYH(664-915)IQ2(ala4)) and normal or five mutated (M149V, E143K, A57G, E56G, R154H) hVLC-1 forms. Double epitope-tagging competition was used to monitor the intracellular localization of exogenously introduced normal and E56G-mutated (hVLC-1(E56G)) hVLC-1 constructs in neonatal rat cardiomyocytes. Fluorescence lifetime imaging microscopy was applied to map the microenvironment of normal and E56G-mutated hVLC-1 in permeabilized muscle fibres. Affinity of M149V, E143K, A57G, and R154H mutated hVLC-1/rß-MYH(664-915)IQ2(ala4) complexes was significantly lower compared with the normal hVLC-1/rß-MYH(664-915)IQ2(ala4) complex interaction. In particular, the E56G mutation induced an ∼30-fold lower MYH affinity. Sorting specificity of E56G-mutated hVLC-1 was negligible compared with normal hVLC-1. Fluorescence lifetime of fibres replaced with hVLC-1(E56G) increased significantly compared with hVLC-1-replaced fibres. CONCLUSION: Disturbed myosin binding of mutated hVLC-1 may provide a pathomechanism for the development of HCM.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Myocytes, Cardiac/physiology , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Sarcomeres/metabolism , Animals , Animals, Newborn , Cells, Cultured , Circular Dichroism , Humans , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Myosin Light Chains/chemistry , Protein Binding/physiology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Surface Plasmon Resonance
13.
J Muscle Res Cell Motil ; 32(4-5): 271-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22057634

ABSTRACT

Ahnak1 is a giant, ubiquitously expressed, plasma membrane support protein whose function in skeletal muscle is largely unknown. Therefore, we investigated whether ahnak would be influenced by alterations of the sarcolemma exemplified by dysferlin mutations known to render the sarcolemma vulnerable or by mutations in calpain3, a protease known to cleave ahnak. Human muscle biopsy specimens obtained from patients with limb girdle muscular dystrophy (LGMD) caused by mutations in dysferlin (LGMD2B) and calpain3 (LGMD2A) were investigated for ahnak expression and localization. We found that ahnak1 has lost its sarcolemmal localization in LGMD2B but not in LGMD2A. Instead ahnak1 appeared in muscle connective tissue surrounding the extracellular site of the muscle fiber in both muscular dystrophies. The entire giant ahnak1 molecule was present outside the muscle fiber and did only partially colocalize with CD45-positive immune cell infiltration and the extracelluar matrix proteins fibronectin and collagenVI. Further, vesicles shedded in response to Ca(2+) by primary human myotubes were purified and their protein content was analysed. Ahnak1 was prominently present in these vesicles. Electron microscopy revealed a homogenous population of vesicles with a diameter of about 150 nm. This is the first study demonstrating vesicle release from human myotubes that may be one mechanism underlying abnormally localized ahnak1. Taken together, our results define ahnak1 in muscle connective tissue as a novel feature of two genetically distinct muscular dystrophies that might contribute to disease pathology.


Subject(s)
Connective Tissue/ultrastructure , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscular Dystrophies, Limb-Girdle/metabolism , Neoplasm Proteins/metabolism , Sarcolemma/ultrastructure , Transport Vesicles/ultrastructure , Calpain/genetics , Calpain/metabolism , Case-Control Studies , Dysferlin , Homozygote , Humans , Immunohistochemistry , Membrane Proteins/genetics , Microscopy, Electron , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , Transport Vesicles/metabolism
14.
J Muscle Res Cell Motil ; 32(4-5): 281-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22038483

ABSTRACT

Ahnak1 has been implicated in the beta-adrenergic regulation of the cardiac L-type Ca(2+) channel current (I (CaL)) by its binding to the regulatory Cavß(2) subunit. In this study, we addressed the question whether ahnak1/Cavß(2) interactions are essential or redundant for beta-adrenergic stimulation of I (CaL). Three naturally occurring ahnak1 variants (V5075 M, G5242R, and T5796 M) identified by genetic screening of cardiomyopathy patients did essentially not influence the in vitro Cavß(2) interaction as assessed by recombinant proteins. But, we observed a robust increase in Cavß(2) binding by mutating Ala at position 4984 to Pro which creates a PxxP consensus motif in the ahnak1 protein fragment. Surface plasmon resonance measurements revealed that this mutation introduced an additional Cavß(2) binding site. The functionality of A4984P was supported by the specific action of the Pro-containing ahnak1-derived peptide (P4984) in beta-adrenergic regulation of I (CaL). Patch clamp recordings on cardiomyocytes showed that intracellular perfusion of P4984 markedly reduced I (CaL) response to the beta-adrenergic agonist, isoprenaline, while the Ala-containing counterpart failed to affect I (CaL). Interestingly, I (CaL) of ahnak1-deficient cardiomyocytes was not affected by peptide application. Moreover, I (CaL) of ahnak1-deficient cardiomyocytes showed intact beta-adrenergic responsiveness. Similarly isolated ahnak1-deficient mouse hearts responded normally to adrenergic challenge. Our results indicate that ahnak1 is not essential for beta-adrenergic up-regulation of I (CaL) and cardiac contractility in mice. But, tuning ahnak1/Cavß(2) interaction provides a tool for modulating the beta-adrenergic response of I (CaL).


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Neoplasm Proteins/metabolism , Adrenergic beta-Agonists/metabolism , Adrenergic beta-Agonists/pharmacology , Amino Acid Motifs , Animals , Binding Sites/physiology , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Signaling/physiology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Case-Control Studies , Humans , Isoproterenol/metabolism , Isoproterenol/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Neoplasm Proteins/genetics , Patch-Clamp Techniques , Polymorphism, Single Nucleotide , Receptors, Adrenergic, beta/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Up-Regulation/drug effects
15.
Hypertension ; 58(1): 77-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21576625

ABSTRACT

Pregnant women who subsequently develop preeclampsia are highly sensitive to infused angiotensin (Ang) II; the sensitivity persists postpartum. Activating autoantibodies against the Ang II type 1 (AT(1)) receptor are present in preeclampsia. In vitro and in vivo data suggest that they could be involved in the disease process. We generated and purified activating antibodies against the AT(1) receptor (AT(1)-AB) by immunizing rabbits against the AFHYESQ epitope of the second extracellular loop, which is the binding epitope of endogenous activating autoantibodies against AT(1) from patients with preeclampsia. We then purified AT(1)-AB using affinity chromatography with the AFHYESQ peptide. We were able to detect AT(1)-AB both by ELISA and a functional bioassay. We then passively transferred AT(1)-AB into pregnant rats, alone or combined with Ang II. AT(1)-AB activated protein kinase C-α and extracellular-related kinase 1/2. Passive transfer of AT(1)-AB alone or Ang II (435 ng/kg per minute) infused alone did not induce a preeclampsia-like syndrome in pregnant rats. However, the combination (AT(1)-AB plus Ang II) induced hypertension, proteinuria, intrauterine growth retardation, and arteriolosclerosis in the uteroplacental unit. We next performed gene-array profiling of the uteroplacental unit and found that hypoxia-inducible factor 1α was upregulated by Ang II plus AT(1)-AB, which we then confirmed by Western blotting in villous explants. Furthermore, endothelin 1 was upregulated in endothelial cells by Ang II plus AT(1)-AB. We show that AT(1)-AB induces Ang II sensitivity. Our mechanistic study supports the existence of an "autoimmune-activating receptor" that could contribute to Ang II sensitivity and possible to preeclampsia.


Subject(s)
Angiotensin II/genetics , Autoantibodies , Gene Expression Regulation, Developmental , Pre-Eclampsia/immunology , Pregnancy, Animal , RNA/genetics , Receptor, Angiotensin, Type 1/immunology , Angiotensin II/metabolism , Animals , Blotting, Western , Cells, Cultured , Cricetinae , Enzyme-Linked Immunosorbent Assay , Female , Fetus/embryology , Fetus/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism
16.
Cardiovasc Res ; 90(3): 513-20, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21262909

ABSTRACT

AIMS: In this paper, we tested the hypothesis that different binding affinities of the C-terminus of human cardiac alkali (essential) myosin light chain (A1) isoforms to the IQ1 motif of the myosin lever arm provide a molecular basis for distinct sarcomeric sorting and inotropic activity. METHODS AND RESULTS: We employed circular dichroism and surface plasmon resonance spectroscopy to investigate structural properties, secondary structures, and protein-protein interactions of a recombinant head-rod fragments of rat cardiac ß-myosin heavy chain aa664-915 with alanine-mutated IQ2 domain (rß-MYH(664-915)IQ(ala4)) and A1 isoforms [human atrial (hALC1) and human ventricular (hVLC-1) light chains]. Double epitope-tagging competition was used to monitor the intracellular localization of exogenously introduced hALC-1 and hVLC-1 constructs in neonatal rat cardiomyocytes. Contractile functions of A1 isoforms were investigated by monitoring shortening and intracellular-free Ca(2+) (Fura-2) of adult rat cardiomyocytes infected with adenoviral (Ad) vectors using hALC-1 or ß-galactosidase as expression cassettes. hALC-1 bound more strongly (greater than three-fold lower K(D)) to rß-MYH(664-915) than did hVLC-1. Sorting specificity of A1 isoforms to sarcomeres of cardiomyocytes rose in the order hVLC-1 to hALC-1. Replacement of endogenous VLC-1 by hALC-1 in adult rat cardiomyocytes increased contractility while the systolic Ca(2+) signal remained unchanged. CONCLUSION: Intense myosin binding of hALC-1 provides a mechanism for preferential sarcomeric sorting and Ca(2+)-independent positive inotropic activity.


Subject(s)
Cardiac Myosins/chemistry , Cardiac Myosins/metabolism , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Amino Acid Substitution , Animals , Animals, Newborn , Atrial Myosins/chemistry , Atrial Myosins/genetics , Atrial Myosins/metabolism , Base Sequence , Calcium Signaling/physiology , Cardiac Myosins/genetics , Circular Dichroism , DNA Primers/genetics , Humans , In Vitro Techniques , Male , Mutagenesis, Site-Directed , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Myosin Light Chains/genetics , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcomeres/metabolism , Surface Plasmon Resonance , Transfection , Ventricular Myosins/chemistry , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
17.
J Biol Chem ; 286(11): 9079-96, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21177871

ABSTRACT

A-kinase anchoring proteins (AKAPs) tether protein kinase A (PKA) and other signaling proteins to defined intracellular sites, thereby establishing compartmentalized cAMP signaling. AKAP-PKA interactions play key roles in various cellular processes, including the regulation of cardiac myocyte contractility. We discovered small molecules, 3,3'-diamino-4,4'-dihydroxydiphenylmethane (FMP-API-1) and its derivatives, which inhibit AKAP-PKA interactions in vitro and in cultured cardiac myocytes. The molecules bind to an allosteric site of regulatory subunits of PKA identifying a hitherto unrecognized region that controls AKAP-PKA interactions. FMP-API-1 also activates PKA. The net effect of FMP-API-1 is a selective interference with compartmentalized cAMP signaling. In cardiac myocytes, FMP-API-1 reveals a novel mechanism involved in terminating ß-adrenoreceptor-induced cAMP synthesis. In addition, FMP-API-1 leads to an increase in contractility of cultured rat cardiac myocytes and intact hearts. Thus, FMP-API-1 represents not only a novel means to study compartmentalized cAMP/PKA signaling but, due to its effects on cardiac myocytes and intact hearts, provides the basis for a new concept in the treatment of chronic heart failure.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase Inhibitors/pharmacology , Second Messenger Systems/physiology , A Kinase Anchor Proteins/genetics , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , Chronic Disease , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Heart Failure/drug therapy , Heart Failure/metabolism , Male , Myocardial Contraction/drug effects , Rats , Rats, Inbred WKY , Second Messenger Systems/drug effects
18.
Biochem Biophys Res Commun ; 401(1): 143-8, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20833135

ABSTRACT

The AHNAK scaffold PDZ-protein family is implicated in various cellular processes including membrane repair; however, AHNAK function and subcellular localization in skeletal muscle are unclear. We used specific AHNAK1 and AHNAK2 antibodies to analyzed the detailed localization of both proteins in mouse skeletal muscle. Co-localization of AHNAK1 and AHNAK2 with vinculin clearly demonstrates that both proteins are components of the costameric network. In contrast, no AHNAK expression was detected in the T-tubule system. A laser wounding assay with AHNAK1-deficient fibers suggests that AHNAK1 is not involved in membrane repair. Using atomic force microscopy (AFM), we observed a significantly higher transverse stiffness of AHNAK1⁻/⁻ fibers. These findings suggest novel functions of AHNAK proteins in skeletal muscle.


Subject(s)
Elastic Modulus , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/chemistry , Neoplasm Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Animals , Cytoskeletal Proteins , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred Strains , Microscopy, Atomic Force , Muscle Fibers, Skeletal/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics
19.
Pflugers Arch ; 460(4): 719-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20607281

ABSTRACT

Ahnak1, a giant 700 kDa protein, has been implicated in Ca(2+) signalling in various cells. Previous work suggested that the interaction between ahnak1 and Cavbeta(2) subunit plays a role in L-type Ca(2+) current (I (CaL)) regulation. Here, we performed structure-function studies with the most C-terminal domain of ahnak1 (188 amino acids) containing a PxxP consensus motif (designated as 188-PSTP) using ventricular cardiomyocytes isolated from rats, wild-type (WT) mice and ahnak1-deficient mice. In vitro binding studies revealed that 188-PSTP conferred high-affinity binding to Cavbeta(2) (K (d) approximately 60 nM). Replacement of proline residues by alanines (188-ASTA) decreased Cavbeta(2) affinity about 20-fold. Both 188-PSTP and 188-ASTA were functional in ahnak1-expressing rat and mouse cardiomyocytes during whole-cell patch clamp. Upon intracellular application, they increased the net Ca(2+) influx by enhancing I (CaL) density and/or increasing I (CaL) inactivation time course without altering voltage dependency. Specifically, 188-ASTA, which failed to affect I (CaL) density, markedly slowed I (CaL) inactivation resulting in a 50-70% increase in transported Ca(2+) during a 0 mV depolarising pulse. Both ahnak1 fragments also slowed current inactivation with Ba(2+) as charge carrier. By contrast, neither 188-PSTP nor 188-ASTA affected any I (CaL) characteristics in ahnak1-deficient mouse cardiomyocytes. Our results indicate that the presence of endogenous ahnak1 is required for tuning the voltage-dependent component of I (CaL) inactivation by ahnak1 fragments. We suggest that ahnak1 modulates the accessibility of molecular determinants in Cavbeta(2) and/or scaffolds selectively different beta-subunit isoforms in the heart.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Neoplasm Proteins/metabolism , Amino Acid Motifs , Animals , Blotting, Western , Calcium Signaling/physiology , Male , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neoplasm Proteins/chemistry , Patch-Clamp Techniques , Rats , Rats, Wistar
20.
Biochem Biophys Res Commun ; 396(4): 939-43, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20460111

ABSTRACT

The denuded IQ2 domain, i.e. myosin heavy chain not associated with regulatory light chains, exerts an inhibitory effect on myosin ATPase activity. In this study, we elaborated a structural explanation for this auto-inhibitory effect of IQ2 on myosin function. We employed analytical ultracentrifugation, circular dichroism, and surface plasmon resonance spectroscopy to investigate structural and functional properties of a myosin heavy chain (MYH) head-rod fragment aa664-915. MYH(664-915) was monomeric, adopted a closed shape, and bound essential myosin light chains (HIS-MLC-1) with low affinity to IQ1. Deletion of IQ2, however opened MYH(664-915). Four amino acids present in IQ2 could be identified to be responsible for this auto-inhibitory structural effect: alanine mutagenesis of I814, Q815, R819, and W827 stretched MYH(664-915) and increased 30-fold the binding affinity of HIS-MLC-1 to IQ1. In this study we show, that denuded IQ2 favours a closed conformation of myosin with a low HIS-MLC-1 binding affinity. The collapsed structure of myosin with denuded IQ2 could explain the auto-inhibitory effects of IQ2 on enzymatic activity of myosin.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Myosin Heavy Chains/chemistry , Myosin Type II/antagonists & inhibitors , Animals , Circular Dichroism , Myosin Heavy Chains/genetics , Myosin Light Chains/chemistry , Protein Structure, Tertiary/genetics , Rats , Sequence Deletion , Surface Plasmon Resonance , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL
...