Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Cells ; 12(9)2023 04 26.
Article in English | MEDLINE | ID: mdl-37174658

ABSTRACT

Plectin, a highly versatile cytolinker protein, is crucial for myofiber integrity and function. Accordingly, mutations in the human gene (PLEC) cause several rare diseases, denoted as plectinopathies, with most of them associated with progressive muscle weakness. Of several plectin isoforms expressed in skeletal muscle and the heart, P1d is the only isoform expressed exclusively in these tissues. Using high-resolution stimulated emission depletion (STED) microscopy, here we show that plectin is located within the gaps between individual α-actinin-positive Z-disks, recruiting and bridging them to desmin intermediate filaments (Ifs). Loss of plectin in myofibril bundles led to a complete loss of desmin Ifs. Loss of Z-disk-associated plectin isoform P1d led to disorganization of muscle fibers and slower relaxation of myofibrils upon mechanical strain, in line with an observed inhomogeneity of muscle ultrastructure. In addition to binding to α-actinin and thereby providing structural support, P1d forms a scaffolding platform for the chaperone-assisted selective autophagy machinery (CASA) by directly interacting with HSC70 and synpo2. In isoform-specific knockout (P1d-KO) mouse muscle and mechanically stretched plectin-deficient myoblasts, we found high levels of undigested filamin C, a bona fide substrate of CASA. Similarly, subjecting P1d-KO mice to forced swim tests led to accumulation of filamin C aggregates in myofibers, highlighting a specific role of P1d in tension-induced proteolysis activated upon high loads of physical exercise and muscle contraction.


Subject(s)
Actinin , Plectin , Animals , Humans , Mice , Desmin/genetics , Desmin/metabolism , Filamins , Plectin/metabolism , Protein Isoforms/metabolism
3.
Front Physiol ; 11: 516, 2020.
Article in English | MEDLINE | ID: mdl-32581830

ABSTRACT

Length-dependent activation of calcium-dependent myocardial force generation provides the basis for the Frank-Starling mechanism. To directly compare the effects of mutations associated with hypertrophic cardiomyopathy and dilated cardiomyopathy, the native troponin complex in skinned trabecular fibers of guinea pigs was exchanged with recombinant heterotrimeric, human, cardiac troponin complexes containing different human cardiac troponin T subunits (hcTnT): hypertrophic cardiomyopathy-associated hcTnTR130C, dilated cardiomyopathy-associated hcTnTΔK210 or the wild type hcTnT (hcTnTWT) serving as control. Force-calcium relations of exchanged fibers were explored at short fiber length defined as 110% of slack length (L 0) and long fiber length defined as 125% of L 0 (1.25 L 0). At short fiber length (1.1 L 0), calcium sensitivity of force generation expressed by -log [Ca2+] required for half-maximum force generation (pCa50) was highest for the hypertrophic cardiomyopathy-associated mutation R130C (5.657 ± 0.019), intermediate for the wild type control (5.580 ± 0.028) and lowest for the dilated cardiomyopathy-associated mutation ΔK210 (5.325 ± 0.038). Lengthening fibers from 1.1 L 0 to 1.25 L 0 increased calcium sensitivity in fibers containing hcTnTR130C (delta-pCa50 = +0.030 ± 0.010), did not alter calcium sensitivity in the wild type control (delta-pCa50 = -0.001 ± 0.010), and decreased calcium sensitivity in fibers containing hcTnTΔK210 (delta-pCa50 = -0.034 ± 0.013). Length-dependent activation indicated by the delta-pCa50 was highly significantly (P < 0.001) different between the two mutations. We hypothesize that primary effects of mutations on length-dependent activation contribute to the development of the diverging phenotypes in hypertrophic and dilated cardiomyopathy.

4.
Front Physiol ; 11: 144, 2020.
Article in English | MEDLINE | ID: mdl-32265723

ABSTRACT

It has been shown that not only calcium but also strong binding myosin heads contribute to thin filament activation in isometrically contracting animal fast-twitch and cardiac muscle preparations. This behavior has not been studied in human muscle fibers or animal slow-twitch fibers. Human slow-twitch fibers are interesting since they contain the same myosin heavy chain isoform as the human heart. To explore myosin-induced activation of the thin filament in isometrically contracting human slow-twitch fibers, the endogenous troponin complex was exchanged for a well-characterized fast-twitch skeletal troponin complex labeled with the fluorescent dye N-((2-(Iodoacetoxy)ethyl)-N-methyl)amino-7-nitrobenz-2-oxa-1,3-diazole (fsTn-IANBD). The exchange was ≈70% complete (n = 8). The relative contributions of calcium and strong binding cross-bridges to thin filament activation were dissected by increasing the concentration of calcium from relaxing (pCa 7.5) to saturating levels (pCa 4.5) before and after incubating the exchanged fibers in the myosin inhibitor para-aminoblebbistatin (AmBleb). At pCa 4.5, the relative contributions of calcium and strong binding cross-bridges to thin filament activation were ≈69 and ≈31%, respectively. Additionally, switching from isometric to isotonic contraction at pCa 4.5 revealed that strong binding cross-bridges contributed ≈29% to thin filament activation (i.e., virtually the same magnitude obtained with AmBleb). Thus, we showed through two different approaches that lowering the number of strong binding cross-bridges, at saturating calcium, significantly reduced the activation of the thin filament in human slow-twitch fibers. The contribution of myosin to activation resembled that which was previously reported in rat cardiac and rabbit fast-twitch muscle preparations. This method could be applied to slow-twitch human fibers obtained from the soleus muscle of cardiomyopathy patients. Such studies could lead to a better understanding of the effect of point mutations of the cardiac myosin head on the regulation of muscle contraction and could lead to better management by pharmacological approaches.

5.
J Cereb Blood Flow Metab ; 37(3): 1014-1029, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27193035

ABSTRACT

Aging causes major alterations of all components of the neurovascular unit and compromises brain blood supply. Here, we tested how aging affects vascular reactivity in basilar arteries from young (<10 weeks; y-BA), old (>22 months; o-BA) and old (>22 months) heterozygous MYPT1-T-696A/+ knock-in mice. In isometrically mounted o-BA, media thickness was increased by ∼10% while the passive length tension relations were not altered. Endothelial denudation or pan-NOS inhibition (100 µmol/L L-NAME) increased the basal tone by 11% in y-BA and 23% in o-BA, while inhibition of nNOS (1 µmol/L L-NPA) induced ∼10% increase in both ages. eNOS expression was ∼2-fold higher in o-BA. In o-BA, U46619-induced force was augmented (pEC50 ∼6.9 vs. pEC50 ∼6.5) while responsiveness to DEA-NONOate, electrical field stimulation or nicotine was decreased. Basal phosphorylation of MLC20-S19 and MYPT1-T-853 was higher in o-BA and was reversed by apocynin. Furthermore, permeabilized o-BA showed enhanced Ca2+-sensitivity. Old T-696A/+ BA displayed a reduced phosphorylation of MYPT1-T696 and MLC20, a lower basal tone in response to L-NAME and a reduced eNOS expression. The results indicate that the vascular hypercontractility found in o-BA is mediated by inhibition of MLCP and is partially compensated by an upregulation of endothelial NO release.


Subject(s)
Acetophenones/pharmacology , Aging , Basilar Artery/physiology , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Animals , Enzyme Inhibitors , Mice , Myosin-Light-Chain Phosphatase/metabolism , Nitric Oxide Synthase Type I/metabolism , Phosphorylation , Protein Subunits/metabolism , Vasoconstriction
6.
Mol Cancer Ther ; 15(5): 971-84, 2016 05.
Article in English | MEDLINE | ID: mdl-26826117

ABSTRACT

The serine/threonine death-associated protein kinases (DAPK) provide pro-death signals in response to (oncogenic) cellular stresses. Lost DAPK expression due to (epi)genetic silencing is found in a broad spectrum of cancers. Within B-cell lymphomas, deficiency of the prototypic family member DAPK1 represents a predisposing or early tumorigenic lesion and high-frequency promoter methylation marks more aggressive diseases. On the basis of protein studies and meta-analyzed gene expression profiling data, we show here that within the low-level context of B-lymphocytic DAPK, particularly CLL cells have lost DAPK1 expression. To target this potential vulnerability, we conceptualized B-cell-specific cytotoxic reconstitution of the DAPK1 tumor suppressor in the format of an immunokinase. After rounds of selections for its most potent cytolytic moiety and optimal ligand part, a DK1KD-SGIII fusion protein containing a constitutive DAPK1 mutant, DK1KD, linked to the scFv SGIII against the B-cell-exclusive endocytic glyco-receptor CD22 was created. Its high purity and large-scale recombinant production provided a stable, selectively binding, and efficiently internalizing construct with preserved robust catalytic activity. DK1KD-SGIII specifically and efficiently killed CD22-positive cells of lymphoma lines and primary CLL samples, sparing healthy donor- or CLL patient-derived non-B cells. The mode of cell death was predominantly PARP-mediated and caspase-dependent conventional apoptosis as well as triggering of an autophagic program. The notoriously high apoptotic threshold of CLL could be overcome by DK1KD-SGIII in vitro also in cases with poor prognostic features, such as therapy resistance. The manufacturing feasibility of the novel CD22-targeting DAPK immunokinase and its selective antileukemic efficiency encourage intensified studies towards specific clinical application. Mol Cancer Ther; 15(5); 971-84. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Death-Associated Protein Kinases/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, B-Cell/metabolism , Recombinant Fusion Proteins/administration & dosage , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Cell Line, Tumor , Death-Associated Protein Kinases/antagonists & inhibitors , Death-Associated Protein Kinases/chemistry , Death-Associated Protein Kinases/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Multigene Family , Mutation , Phosphorylation , Protein Interaction Domains and Motifs/genetics , Single-Chain Antibodies/administration & dosage
7.
Proc Natl Acad Sci U S A ; 113(2): E155-64, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26715748

ABSTRACT

Autoimmune response to cardiac troponin I (TnI) induces inflammation and fibrosis in the myocardium. High-mobility group box 1 (HMGB1) is a multifunctional protein that exerts proinflammatory activity by mainly binding to receptor for advanced glycation end products (RAGE). The involvement of the HMGB1-RAGE axis in the pathogenesis of inflammatory cardiomyopathy is yet not fully understood. Using the well-established model of TnI-induced experimental autoimmune myocarditis (EAM), we demonstrated that both local and systemic HMGB1 protein expression was elevated in wild-type (wt) mice after TnI immunization. Additionally, pharmacological inhibition of HMGB1 using glycyrrhizin or anti-HMGB1 antibody reduced inflammation in hearts of TnI-immunized wt mice. Furthermore, RAGE knockout (RAGE-ko) mice immunized with TnI showed no structural or physiological signs of cardiac impairment. Moreover, cardiac overexpression of HMGB1 using adeno-associated virus (AAV) vectors induced inflammation in the hearts of both wt and RAGE-ko mice. Finally, patients with myocarditis displayed increased local and systemic HMGB1 and soluble RAGE (sRAGE) expression. Together, our study highlights that HMGB1 and its main receptor, RAGE, appear to be crucial factors in the pathogenesis of TnI-induced EAM, because inhibition of HMGB1 and ablation of RAGE suppressed inflammation in the heart. Moreover, the proinflammatory effect of HMGB1 is not necessarily dependent on RAGE only. Other receptors of HMGB1 such as Toll-like receptors (TLRs) may also be involved in disease pathogenesis. These findings could be confirmed by the clinical relevance of HMGB1 and sRAGE. Therefore, blockage of one of these molecules might represent a novel therapeutic strategy in the treatment of autoimmune myocarditis and inflammatory cardiomyopathy.


Subject(s)
HMGB1 Protein/metabolism , Inflammation/genetics , Receptor for Advanced Glycation End Products/metabolism , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Biopsy , Dependovirus/metabolism , Down-Regulation/drug effects , Fibrosis , Freund's Adjuvant/immunology , HMGB1 Protein/blood , Heart Diseases/blood , Heart Diseases/complications , Heart Diseases/genetics , Heart Diseases/pathology , Heart Function Tests , Immunization , Immunologic Factors/pharmacology , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Matrix Metalloproteinases/metabolism , Mice, Knockout , Myocarditis/complications , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/metabolism , Protein Binding/drug effects , Receptor for Advanced Glycation End Products/blood , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Transcription, Genetic/drug effects , Troponin/metabolism , Up-Regulation/drug effects
8.
Biophys J ; 103(6): 1254-64, 2012 Sep 19.
Article in English | MEDLINE | ID: mdl-22995498

ABSTRACT

Conformational changes in the skeletal troponin complex (sTn) induced by rapidly increasing or decreasing the [Ca(2+)] were probed by 5-iodoacetamidofluorescein covalently bound to Cys-133 of skeletal troponin I (sTnI). Kinetics of conformational changes was determined for the isolated complex and after incorporating the complex into rabbit psoas myofibrils. Isolated and incorporated sTn exhibited biphasic Ca(2+)-activation kinetics. Whereas the fast phase (k(obs)∼1000 s(-1)) is only observed in this study, where kinetics were induced by Ca(2+), the slower phase resembles the monophasic kinetics of sTnI switching observed in another study (Brenner and Chalovich. 1999. Biophys. J. 77:2692-2708) that investigated the sTnI switching induced by releasing the feedback of force-generating cross-bridges on thin filament activation. Therefore, the slower conformational change likely reflects the sTnI switch that regulates force development. Modeling reveals that the fast conformational change can occur after the first Ca(2+) ion binds to skeletal troponin C (sTnC), whereas the slower change requires Ca(2+) binding to both regulatory sites of sTnC. Incorporating sTn into myofibrils increased the off-rate and lowered the Ca(2+) sensitivity of sTnI switching. Comparison of switch-off kinetics with myofibril force relaxation kinetics measured in a mechanical setup indicates that sTnI switching might limit the rate of fast skeletal muscle relaxation.


Subject(s)
Calcium/pharmacology , Myofibrils/drug effects , Myofibrils/metabolism , Troponin I/metabolism , Animals , Biomechanical Phenomena , Fluoresceins/metabolism , In Vitro Techniques , Kinetics , Muscle Relaxation/drug effects , Myofibrils/physiology , Psoas Muscles/drug effects , Rabbits , Sarcomeres/drug effects , Sarcomeres/metabolism
9.
J Am Heart Assoc ; 1(6): e003293, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23316321

ABSTRACT

BACKGROUND: Overexpression of interleukin-10 (IL-10) in murine CD11b(+) monocytes/macrophages via GMP-adapted mRNA-nucleofection was expected to improve clinical outcome and reduce adverse side effects in autoimmune myocarditis. This study represents the proof of principle for a novel anti-inflammatory therapy using overexpression of IL-10 in murine monocytes/macrophages by mRNA-nucleofection for the treatment of autoimmune myocarditis. METHODS AND RESULTS: Autoimmune myocarditis was induced in A/J mice by subcutaneous immunization with troponin I. CD11b(+) monocytes/macrophages were isolated from the peritoneum and IL-10 was overexpressed by mRNA-nucleofection. These cells were injected intravenously. Myocardial inflammation was assessed via histological and immunohistochemical examinations. Myocardial fibrosis was analyzed with Masson's trichrome staining. Antitroponin I antibodies were determined within the serum. Physical performance was evaluated using a running wheel and echocardiography. In vitro overexpression of IL-10 in CD11b(+) monocytes/macrophages resulted in a 7-fold increased production of IL-10 (n=3). In vivo higher levels of IL-10 and less inflammation were detected within the myocardium of treated compared with control mice (n=4). IL-10-treated mice showed lower antitroponin I antibodies (n=10) and a better physical performance (n=10). CONCLUSIONS: Application of IL-10-overexpressing CD11b(+) monocytes/macrophages reduced inflammation and improved physical performance in a murine model of autoimmune myocarditis. Thus, the use of genetically modified monocytes/macrophages facilitated a targeted therapy of local inflammation and may reduce systemic side effects. Because the nucleofection technique is GMP adapted, an in vivo use in humans seems basically feasible and the transfer to other inflammatory diseases seems likely.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/therapy , Interleukin-10/therapeutic use , Myocarditis/therapy , Animals , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/metabolism , Autoimmune Diseases/metabolism , Autoimmune Diseases/physiopathology , Cell Movement , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Feasibility Studies , Female , Gene Expression , Humans , Inflammation/metabolism , Inflammation/physiopathology , Inflammation/therapy , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-10/metabolism , Macrophages/metabolism , Macrophages/physiology , Mice , Monocytes/metabolism , Monocytes/physiology , Myocarditis/metabolism , Myocarditis/physiopathology , RNA, Messenger , Transfection
10.
Circ Res ; 109(2): 130-40, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21597011

ABSTRACT

RATIONALE: This study was performed to gain insights into novel therapeutic approaches for the treatment of autoimmune myocarditis. OBJECTIVE: Chemical stimulation of the efferent arm of the vagus nerve through activation of nicotinic acetylcholine receptor subtype-7α (α7-nAChR) has been shown to be protective in several models of inflammatory diseases. In the present study, we investigated the potentially protective effect of vagus nerve stimulation on myocarditis. METHODS AND RESULTS: A/J mice were immunized with cardiac troponin I (TnI) to induce autoimmune myocarditis. Mice were exposed to drinking water that contained nicotine in different concentrations and for different time periods (for 3 days at 12.5 mg/L; 3 days at 125 mg/L; 21 days at 12.5 mg/L; and 21 days at 125 mg/L after first immunization). TnI-immunized mice with no pharmacological treatment showed extensive myocardial inflammation and fibrosis and significantly elevated levels of interleukin-6 and tumor necrosis factor-α. Furthermore, elevated levels of mRNA transcripts of proinflammatory chemokines (monocyte chemoattractant protein-1, macrophage inflammatory protein-1ß, and RANTES) and chemokine receptors (CCR1, CCR2, and CCR5) were found. Oral nicotine administration reduced inflammation within the myocardium, decreased the production of interleukin-6 and tumor necrosis factor-α, and downregulated the expression of monocyte chemoattractant protein-1, macrophage inflammatory protein-1ß, RANTES, CCR1, CCR2, and CCR5. In addition, nicotine treatment resulted in decreased expression of matrix metalloproteinase-14, natriuretic peptide precursor B, tissue inhibitor of metalloproteinase-1, and osteopontin, proteins that are commonly involved in heart failure. Finally, we found that nicotine reduced levels of pSTAT3 (phosphorylated signal transducer and activator of transcription 3) protein expression within the myocardium. Neostigmine treatment did not affect the progression of myocarditis. CONCLUSIONS: We showed that activation of the cholinergic antiinflammatory pathway with nicotine reduces inflammation in autoimmune myocarditis. Our results may open new possibilities in the therapeutic management of autoimmune myocarditis.


Subject(s)
Cholinergic Agents/immunology , Myocarditis/etiology , Nicotine/pharmacology , Animals , Autoimmune Diseases/etiology , Cytokines/analysis , Cytokines/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Inflammation , Inflammation Mediators , Mice , Nicotine/administration & dosage , Nicotine/therapeutic use , Proteins/analysis , RNA, Messenger/analysis , Troponin I , Vagus Nerve
11.
Biochem J ; 429(2): 291-302, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20459395

ABSTRACT

KRP (kinase-related protein), also known as telokin, has been proposed to inhibit smooth muscle contractility by inhibiting the phosphorylation of the rMLC (regulatory myosin light chain) by the Ca2+-activated MLCK (myosin light chain kinase). Using the phosphatase inhibitor microcystin, we show in the present study that KRP also inhibits Ca2+-independent rMLC phosphorylation and smooth muscle contraction mediated by novel Ca2+-independent rMLC kinases. Incubating KRP-depleted Triton-skinned taenia coli with microcystin at pCa>8 induced a slow contraction reaching 90% of maximal force (Fmax) at pCa 4.5 after approximately 25 min. Loading the fibres with KRP significantly slowed down the force development, i.e. the time to reach 50% of Fmax was increased from 8 min to 35 min. KRP similarly inhibited rMLC phosphorylation of HMM (heavy meromyosin) in vitro by MLCK or by the constitutively active MLCK fragment (61K-MLCK) lacking the myosin-docking KRP domain. A C-terminally truncated KRP defective in myosin binding inhibited neither force nor HMM phosphorylation. Phosphorylated KRP inhibited the rMLC phosphorylation of HMM in vitro and Ca2+-insensitive contractions in fibres similar to unphosphorylated KRP, whereby the phosphorylation state of KRP was not altered in the fibres. We conclude that (i) KRP inhibits not only MLCK-induced contractions, but also those elicited by Ca2+-independent rMLC kinases; (ii) phosphorylation of KRP does not modulate this effect; (iii) binding of KRP to myosin is essential for this inhibition; and (iv) KRP inhibition of rMLC phosphorylation is most probably due to the shielding of the phosphorylation site on the rMLC.


Subject(s)
Colon/physiology , Muscle Contraction/physiology , Myosin-Light-Chain Kinase/metabolism , Peptide Fragments/metabolism , Animals , Base Sequence , Binding Sites , Calcium/metabolism , Calcium/pharmacology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , Chickens , Colon/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Primers/genetics , Female , Guinea Pigs , Humans , In Vitro Techniques , Male , Marine Toxins , Microcystins/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Myosin Light Chains/chemistry , Myosin Light Chains/metabolism , Myosin Subfragments/chemistry , Myosin Subfragments/metabolism , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/pharmacology , Octoxynol , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Phosphorylation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
12.
Circulation ; 118(20): 2063-72, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18955666

ABSTRACT

BACKGROUND: Despite the widespread use of cardiac troponins for diagnosis of myocyte injury and risk stratification in acute cardiac disorders, little is known about the long-term effects of the released troponins on cardiac function. Recently, we showed that an autoimmune response to cardiac troponin I (cTnI) induces severe inflammation and subsequent fibrosis in the myocardium. This autoimmune disorder predisposes to heart failure and cardiac death in mice. METHODS AND RESULTS: To investigate the role of cTnI-specific T cells, T cells were isolated from splenocytes of mice immunized with murine cTnI (mcTnI). Wild-type mice that received mcTnI-specific T cells showed high mcTnI-specific antibody titers, increased production of the proinflammatory cytokines interleukin-1beta and tumor necrosis factor-alpha, severe inflammation and fibrosis in the myocardium, and reduced fractional shortening. To identify the antigenic determinants of troponin I responsible for the observed inflammation, fibrosis, and heart failure, 16 overlapping 16mer to 18mer peptides covering the entire amino acid sequence of mcTnI (211 residues) were synthesized. Only mice immunized with residues 105 to 122 of mcTnI developed significant inflammation and fibrosis in the myocardium, with increased expression of the inflammatory chemokines RANTES, monocyte chemotactic protein-1, macrophage inflammatory protein-1alpha, macrophage inflammatory protein-1beta, macrophage inflammatory protein-2, T-cell activation-3, and eotaxin and the chemokine receptors CCR1, CCR2, and CCR5. Mice immunized with the corresponding human cTnI residues 104 to 121 and the mcTnI residues 131 to 148 developed milder disease. CONCLUSIONS: Transfer of troponin I-specific T cells can induce inflammation and fibrosis in wild-type mice, which leads to deterioration of contractile function. Furthermore, 2 sequence motifs of cTnI that induce inflammation and fibrosis in the myocardium are characterized.


Subject(s)
Heart Failure/genetics , Myocarditis/genetics , Myocardium/metabolism , Myocardium/pathology , Troponin I/genetics , Troponin I/immunology , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Antibody Formation , Autoimmunity , Bone and Bones/metabolism , Chemokines/metabolism , Female , Fibrosis , Humans , Immunization , Mice , Mice, Inbred Strains , Mice, SCID , Molecular Conformation , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Peptide Fragments/immunology , Receptors, Chemokine/metabolism , Sequence Analysis, Protein , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Troponin I/chemistry , Troponin I/metabolism
13.
Biophys J ; 93(11): 3917-31, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17704185

ABSTRACT

The kinetics of Ca(2+)-dependent conformational changes of human cardiac troponin (cTn) were studied on isolated cTn and within the sarcomeric environment of myofibrils. Human cTnC was selectively labeled on cysteine 84 with N-((2-(iodoacetoxy)ethyl)-N-methyl)amino-7-nitrobenz-2-oxa-1,3-diazole and reconstituted with cTnI and cTnT to the cTn complex, which was incorporated into guinea pig cardiac myofibrils. These exchanged myofibrils, or the isolated cTn, were rapidly mixed in a stopped-flow apparatus with different [Ca(2+)] or the Ca(2+)-buffer 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid to determine the kinetics of the switch-on or switch-off, respectively, of cTn. Activation of myofibrils with high [Ca(2+)] (pCa 4.6) induced a biphasic fluorescence increase with rate constants of >2000 s(-1) and approximately 330 s(-1), respectively. At low [Ca(2+)] (pCa 6.6), the slower rate was reduced to approximately 25 s(-1), but was still approximately 50-fold higher than the rate constant of Ca(2+)-induced myofibrillar force development measured in a mechanical setup. Decreasing [Ca(2+)] from pCa 5.0-7.9 induced a fluorescence decay with a rate constant of 39 s(-1), which was approximately fivefold faster than force relaxation. Modeling the data indicates two sequentially coupled conformational changes of cTnC in myofibrils: 1), rapid Ca(2+)-binding (k(B) approximately 120 microM(-1) s(-1)) and dissociation (k(D) approximately 550 s(-1)); and 2), slower switch-on (k(on) = 390s(-1)) and switch-off (k(off) = 36s(-1)) kinetics. At high [Ca(2+)], approximately 90% of cTnC is switched on. Both switch-on and switch-off kinetics of incorporated cTn were around fourfold faster than those of isolated cTn. In conclusion, the switch kinetics of cTn are sensitively changed by its structural integration in the sarcomere and directly rate-limit neither cardiac myofibrillar contraction nor relaxation.


Subject(s)
Calcium/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Myofibrils/physiology , Troponin/metabolism , Animals , Cells, Cultured , Female , Guinea Pigs , Humans , Kinetics
14.
Cardiovasc Res ; 74(1): 124-32, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17321507

ABSTRACT

OBJECTIVE: Phosphorylation of proteins in cardiac myofilaments is a major determinant in the regulation of the Ca(2+) sensitivity of contraction. Whereas most reports have focused on effects of phosphorylation, little is known about reverse effects of dephosphorylation in skinned myocardium. Here we studied the effect of the Mn(2+)-dependent catalytic subunit of protein phosphatase 1 (PP1c-alpha) on the Ca(2+) regulation of contraction. In particular, we tested the hypothesis that phosphorylation persists after the skinning procedure and thereby attenuates protein kinase A (PKA)-induced Ca(2+) desensitisation. METHODS: Effects of Mn(2+) and Mn(2+)-PP1c on the Ca(2+) sensitivity of contraction (pCa(50)) were investigated in triton-skinned cardiac fibres from mice and compared with those of PKA treatment. Phosphorylation of proteins was monitored by autoradiography. RESULTS: PKA treatment significantly decreased the pCa(50) by 0.04 pCa units. In contrast, treatment with PP1c or Mn(2+)-containing PP1c buffer significantly increased the pCa(50) by 0.26 units or 0.09 units, respectively. These Ca(2+) sensitisations were completely reversed by subsequent PKA treatment. Replacement of the endogenous cardiac troponin I (cTnI) in fibres with the phospho-mimicking mutant human cTnI(S22/23D) abolished the PP1c-induced Ca(2+) sensitisation. PP1c removed (32)P which had been incorporated into cTnI and cardiac myosin binding protein C by PKA treatment. PKA incorporated twofold more (32)P into cTnI in fibres pre-treated with PP1c. CONCLUSIONS: Mn(2+)-dependent PP1c increases the Ca(2+) sensitivity of contraction of skinned cardiac fibres. This can be ascribed to dephosphorylation of PKA-dependent phosphorylation sites. Hence PKA-dependent phosphorylation of sarcomeric proteins persists to a functionally relevant degree after the skinning procedure.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/pharmacology , Manganese/pharmacology , Myocardium/metabolism , Phosphoprotein Phosphatases/pharmacology , Animals , Autoradiography , Blotting, Western/methods , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , In Vitro Techniques , Mice , Models, Animal , Myocardial Contraction/drug effects , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Phosphatase 1 , Sarcomeres/metabolism , Troponin I/analysis , Troponin I/metabolism
15.
Circulation ; 114(16): 1693-702, 2006 Oct 17.
Article in English | MEDLINE | ID: mdl-17015788

ABSTRACT

BACKGROUND: Cardiac troponins in blood are the most preferred markers of myocardial damage. The fact that they are normally not found in the circulation provides a high level of clinical sensitivity and specificity even when cardiac lesions are small. After myocardial injury, the troponins enter the circulation, where they can be used for diagnosis of acute coronary syndromes. Thus, the cardiac troponins are paramount for disease classification and risk stratification. However, little is known about the long-term effects of the released troponins on cardiac function. METHODS AND RESULTS: In this study we prepared recombinant murine cardiac troponin I (mc-TnI) and murine cardiac troponin T and used them to immunize mice. We report that A/J mice immunized with mc-TnI developed severe inflammation of the myocardium with increased expression of inflammatory chemokines RANTES (regulated on activation normal T cell expressed and secreted), monocyte chemoattractant protein-1, macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, MIP-2, T-cell activation gene 3, and eotaxin and chemokine receptors CCR1, CCR2, and CCR5. The inflammation was followed by cardiomegaly, fibrosis, reduced fractional shortening, and 30% mortality over 270 days. In contrast, mice immunized with murine cardiac troponin T or with the control buffer showed little or no inflammation and no death. Furthermore, we demonstrate that mice preimmunized with mc-TnI before left anterior descending coronary artery ligation showed greater infarct size, more fibrosis, higher inflammation score, and reduced fractional shortening. CONCLUSIONS: Overall, our results show for the first time that provocation of an autoimmune response to mc-TnI induces severe inflammation in the myocardium followed by fibrosis and heart failure with increased mortality in mice.


Subject(s)
Autoimmune Diseases/immunology , Myocarditis/immunology , Myocardium/immunology , Troponin I/immunology , Troponin T/immunology , Animals , Female , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Troponin I/toxicity , Troponin T/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...