Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 108: 127-137, 2017 07.
Article in English | MEDLINE | ID: mdl-28600229

ABSTRACT

Recently, linkage analysis of two large unrelated multigenerational families identified a novel dilated cardiomyopathy (DCM)-linked mutation in the gene coding for alpha-tropomyosin (TPM1) resulting in the substitution of an aspartic acid for an asparagine (at residue 230). To determine how a single amino acid mutation in α-tropomyosin (Tm) can lead to a highly penetrant DCM we generated a novel transgenic mouse model carrying the D230N mutation. The resultant mouse model strongly phenocopied the early onset of cardiomyopathic remodeling observed in patients as significant systolic dysfunction was observed by 2months of age. To determine the precise cellular mechanism(s) leading to the observed cardiac pathology we examined the effect of the mutation on Ca2+ handling in isolated myocytes and myofilament activation in vitro. D230N-Tm filaments exhibited a reduced Ca2+ sensitivity of sliding velocity. This decrease in sensitivity was coupled to increase in the peak amplitude of Ca2+ transients. While significant, and consistent with other DCMs, these measurements are comprised of complex inputs and did not provide sufficient experimental resolution. We then assessed the primary structural effects of D230N-Tm. Measurements of the thermal unfolding of D230N-Tm vs WT-Tm revealed an increase in stability primarily affecting the C-terminus of the Tm coiled-coil. We conclude that the D230N-Tm mutation induces a decrease in flexibility of the C-terminus via propagation through the helical structure of the protein, thus decreasing the flexibility of the Tm overlap and impairing its ability to regulate contraction. Understanding this unique structural mechanism could provide novel targets for eventual therapeutic interventions in patients with Tm-linked cardiomyopathies.


Subject(s)
Amino Acid Substitution , Cardiomyopathy, Dilated/genetics , Mutation , Tropomyosin/chemistry , Tropomyosin/genetics , Animals , Calcium/chemistry , Calcium/metabolism , Cardiomyopathy, Dilated/diagnostic imaging , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Codon , Disease Models, Animal , Echocardiography , Gene Expression , Heart Function Tests , Humans , Mice , Mice, Transgenic , Models, Molecular , Myofibrils , Myosins/genetics , Myosins/metabolism , Protein Conformation , Protein Stability , Structure-Activity Relationship , Thermodynamics , Tropomyosin/metabolism , Troponin/genetics , Troponin/metabolism
2.
J Mol Cell Cardiol ; 91: 42-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26714042

ABSTRACT

Abnormalities of cardiomyocyte Ca(2+) homeostasis and excitation-contraction (E-C) coupling are early events in the pathogenesis of hypertrophic cardiomyopathy (HCM) and concomitant determinants of the diastolic dysfunction and arrhythmias typical of the disease. T-tubule remodelling has been reported to occur in HCM but little is known about its role in the E-C coupling alterations of HCM. Here, the role of T-tubule remodelling in the electro-mechanical dysfunction associated to HCM is investigated in the Δ160E cTnT mouse model that expresses a clinically-relevant HCM mutation. Contractile function of intact ventricular trabeculae is assessed in Δ160E mice and wild-type siblings. As compared with wild-type, Δ160E trabeculae show prolonged kinetics of force development and relaxation, blunted force-frequency response with reduced active tension at high stimulation frequency, and increased occurrence of spontaneous contractions. Consistently, prolonged Ca(2+) transient in terms of rise and duration are also observed in Δ160E trabeculae and isolated cardiomyocytes. Confocal imaging in cells isolated from Δ160E mice reveals significant, though modest, remodelling of T-tubular architecture. A two-photon random access microscope is employed to dissect the spatio-temporal relationship between T-tubular electrical activity and local Ca(2+) release in isolated cardiomyocytes. In Δ160E cardiomyocytes, a significant number of T-tubules (>20%) fails to propagate action potentials, with consequent delay of local Ca(2+) release. At variance with wild-type, we also observe significantly increased variability of local Ca(2+) transient rise as well as higher Ca(2+)-spark frequency. Although T-tubule structural remodelling in Δ160E myocytes is modest, T-tubule functional defects determine non-homogeneous Ca(2+) release and delayed myofilament activation that significantly contribute to mechanical dysfunction.


Subject(s)
Cardiomyopathy, Hypertrophic/physiopathology , Excitation Contraction Coupling , Myocardial Contraction , Myocytes, Cardiac/pathology , Myofibrils/pathology , Sarcolemma/pathology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Actin Cytoskeleton/ultrastructure , Action Potentials , Animals , Calcium/metabolism , Calcium Signaling , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Disease Models, Animal , Gene Expression , Humans , Ion Transport , Mice , Mice, Knockout , Microscopy, Confocal , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Myofibrils/metabolism , Myofibrils/ultrastructure , Optical Imaging , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Troponin T/genetics , Troponin T/metabolism
3.
J Physiol ; 536(Pt 2): 583-92, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11600691

ABSTRACT

1. The heterogenic nature of familial hypertrophic cardiomyopathy (FHC) in humans suggests a link between the type of mutation and the nature of patho-physiological alterations in cardiac myocytes. Exactly how FHC-associated mutations in cardiac troponin T (cTnT) lead to impaired cardiac function is unclear. 2. We measured steady-state isometric force and ATPase activity in detergent-skinned cardiac fibre bundles from three transgenic (TG) mouse hearts in which 50, 92 and 6 % of the native cTnT was replaced by the wild type (WT) cTnT, R92Q mutant cTnT (R92Q) and the C-terminal deletion mutant of cTnT (cTnT(DEL)), respectively. 3. Normalized pCa-tension relationships of R92Q and cTnT(DEL) fibres demonstrated a significant increase in sensitivity to Ca2+ at short (2.0 microm) and long (2.3 microm) sarcomere lengths (SL). At short SL, the pCa50 values, representing the midpoint of the pCa-tension relationship, were 5.69 +/- 0.01, 5.96 +/- 0.01 and 5.81 +/- 0.01 for WT, R92Q and cTnT(DEL) fibres, respectively. At long SL, the pCa50 values were 5.81 +/- 0.01, 6.08 +/- 0.01 and 5.95 +/- 0.01 for WT, R92Q and cTnT(DEL) fibres, respectively. 4. The difference in pCa required for half-maximal activation (DeltapCa50) at short and long SL was 0.12 +/- 0.01 for the R92Q (92 %) TG fibres, which is significantly less than the previously reported DeltapCa50 value of 0.29 +/- 0.02 for R92Q (67 %) TG fibres. 5. At short SL, Ca2+-activated maximal tension in both R92Q and cTnT(DEL) fibres decreased significantly (24 and 21 %, respectively; P < 0.005), with no corresponding decrease in Ca2+-activated maximal ATPase activity. Therefore, at short SL, the tension cost in R92Q and cTnT(DEL) fibres increased by 35 and 29 %, respectively (P < 0.001). 6. The fibre bundles reconstituted with the recombinant mutant cTnT(DEL) protein developed only 37 % of the Ca2+-activated maximal force developed by recombinant WT cTnT reconstituted fibre bundles, with no apparent changes in Ca2+ sensitivity. 7. Our data indicate that an important mutation-linked effect on cardiac function is the result of an inefficient use of ATP at the myofilament level. Furthermore, the extent of the mutation-induced dysfunction depends not only on the nature of the mutation, but also on the concentration of the mutant protein in the sarcomere.


Subject(s)
Actin Cytoskeleton/physiology , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Troponin T/genetics , Animals , Calcium/metabolism , Detergents , Energy Metabolism/physiology , In Vitro Techniques , Mice , Mice, Transgenic , Mutation/physiology , Myocardial Contraction/physiology , Myocardium/cytology , Myocardium/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Troponin T/metabolism
4.
Am J Physiol Heart Circ Physiol ; 280(2): H705-13, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158969

ABSTRACT

The functional consequences of the R92Q mutation in cardiac troponin T (cTnT), linked to familial hypertrophic cardiomyopathy in humans, are not well understood. We have studied steady- and pre-steady-state mechanical activity of detergent-skinned fiber bundles from a transgenic (TG) mouse model in which 67% of the total cTnT in the heart was replaced by the R92Q mutant cTnT. TG fibers were more sensitive to Ca(2+) than nontransgenic (NTG) fibers [negative logarithm of half maximally activating molar Ca(2+) (pCa(50)) = 5.84 +/- 0.01 and 6.12 +/- 0.01 for NTG and TG fibers, respectively]. The shift in pCa(50) caused by increasing the sarcomere length from 1.9 to 2.3 microm was significantly higher for TG than for NTG fibers (DeltapCa(50) = 0.13 +/- 0.01 and 0.29 +/- 0.02 for NTG and TG fibers, respectively). The relationships between rate of ATP consumption and steady-state isometric tension were linear, and the slopes were the same in NTG and TG fibers. Rate of tension redevelopment was more sensitive to Ca(2+) in TG than in NTG fibers (pCa(50) = 5.71 +/- 0.02 and 6.07 +/- 0.02 for NTG and TG fibers, respectively). We concluded that overall cross-bridge cycling kinetics are not altered by the R92Q mutation but that altered troponin-tropomyosin interactions could be responsible for the increase in myofilament Ca(2+) sensitivity in TG myofilaments.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Muscle Fibers, Skeletal/physiology , Troponin T/genetics , Troponin T/metabolism , Acidosis/physiopathology , Adenosine Triphosphate/metabolism , Animals , Calcium/pharmacology , Cross-Linking Reagents/metabolism , Detergents , Genes, myc/genetics , Humans , Mice , Mice, Transgenic , Muscle Contraction/drug effects , Muscle Contraction/physiology , Mutation/physiology , Promoter Regions, Genetic/physiology
5.
Am J Physiol Heart Circ Physiol ; 278(2): H412-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666070

ABSTRACT

Alpha- and beta-myosin heavy chain (MHC), the two MHC isoforms expressed in the mammalian heart, differ quantitatively in their enzymatic activities. The MHC composition of the heart can change dramatically in response to numerous stimuli, leading to the hypothesis that changes in cardiac function can be caused by myosin isoform shifts. However, this hypothesis has remained unproven because the stimuli used to generate these shifts are complex and accompanied by many additional physiological changes, including alterations in cardiac mass and geometry. Adult mouse ventricles normally express only alpha-MHC (the faster motor). To determine whether genetic alteration of the MHC isoform composition in the adult mouse heart would result in changes in cardiac chamber mass and contractility, we established transgenic mouse lines that express a Myc-tagged beta-MHC molecule (the slower motor) in adult ventricular tissue, one of which expresses 12% of its myosin as the transgene. There is no evidence of hypertrophy, induction of hypertrophic markers, and no histopathology. Myofibrillar Ca(2+)-activated ATPase activity is decreased by 23%, and Langendorff preparations demonstrate a significant 15% decrease in systolic function in transgenic hearts. These results suggest that even small shifts in the myosin isoform composition of the myocardium can result in physiologically significant changes in cardiac contractility and could be relevant to cardiovascular disease.


Subject(s)
Genes, Dominant/physiology , Heart/physiology , Myocardium/metabolism , Myosin Heavy Chains/metabolism , Animals , Calcium-Transporting ATPases/metabolism , Gene Expression , Genes, myc/genetics , Heart/anatomy & histology , Mice , Mice, Transgenic/genetics , Myocardial Contraction/physiology , Myofibrils/enzymology , Myosin Heavy Chains/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Sequence Tagged Sites , Transgenes/genetics
6.
J Clin Invest ; 104(4): 469-81, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10449439

ABSTRACT

Multiple mutations in cardiac troponin T (cTnT) can cause familial hypertrophic cardiomyopathy (FHC). Patients with cTnT mutations generally exhibit mild or no ventricular hypertrophy, yet demonstrate a high frequency of early sudden death. To understand the functional basis of these phenotypes, we created transgenic mouse lines expressing 30%, 67%, and 92% of their total cTnT as a missense (R92Q) allele analogous to one found in FHC. Similar to a mouse FHC model expressing a truncated cTnT protein, the left ventricles of all R92Q lines are smaller than those of wild-type. In striking contrast to truncation mice, however, the R92Q hearts demonstrate significant induction of atrial natriuretic factor and beta-myosin heavy chain transcripts, interstitial fibrosis, and mitochondrial pathology. Isolated cardiac myocytes from R92Q mice have increased basal sarcomeric activation, impaired relaxation, and shorter sarcomere lengths. Isolated working heart data are consistent, showing hypercontractility and diastolic dysfunction, both of which are common findings in patients with FHC. These mice represent the first disease model to exhibit hypercontractility, as well as a unique model system for exploring the cellular pathogenesis of FHC. The distinct phenotypes of mice with different TnT alleles suggest that the clinical heterogeneity of FHC is at least partially due to allele-specific mechanisms.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Mutation, Missense , Troponin T/genetics , Alleles , Animals , Atrial Natriuretic Factor/genetics , Base Sequence , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Size , DNA Primers/genetics , Disease Models, Animal , Heart Ventricles/pathology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microscopy, Electron , Myocardial Contraction , Myosin Heavy Chains/genetics , Phenotype , Sarcomeres/ultrastructure
7.
Nucleic Acids Res ; 26(16): 3794-9, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9685498

ABSTRACT

The alpha1-protease inhibitor proteins of laboratory mice are homologous in sequence and function to human alpha1-antitrypsin and are encoded by a highly conserved multigene family comprised of five members. In humans, the inhibitor is expressed in liver and in macrophages and decreased expression or inhibitory activity is associated with a deficiency syndrome which can result in emphysema and liver disease in affected individuals. It has been proposed that macrophage expression may be an important component of the function of human alpha1-antitrypsin. Clearly, it is desirable to develop a mouse model of this deficiency syndrome, however, efforts to do this have been largely unsuccessful. In this paper, we report that aside from the issues of potentially redundant gene function, the mouse may not be a suitable animal for such studies, because there is no significant expression of murine alpha1-protease inhibitor in the macrophages of mice. This difference between the species appears to result from an absence of a functional macrophage-specific promoter in mice.


Subject(s)
alpha 1-Antitrypsin/genetics , Animals , Base Sequence , Conserved Sequence , DNA/genetics , DNA Primers/genetics , Disease Models, Animal , Gene Expression , Humans , Liver/metabolism , Macrophages/metabolism , Mice , Multigene Family , Polymerase Chain Reaction , Promoter Regions, Genetic , Sequence Homology, Nucleic Acid , Species Specificity , Syndrome , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin Deficiency/metabolism
8.
J Clin Invest ; 101(12): 2800-11, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9637714

ABSTRACT

Mutations in multiple cardiac sarcomeric proteins including myosin heavy chain (MyHC) and cardiac troponin T (cTnT) cause a dominant genetic heart disease, familial hypertrophic cardiomyopathy (FHC). Patients with mutations in these two genes have quite distinct clinical characteristics. Those with MyHC mutations demonstrate more significant and uniform cardiac hypertrophy and a variable frequency of sudden death. Patients with cTnT mutations generally exhibit mild or no hypertrophy, but a high frequency of sudden death at an early age. To understand the basis for these distinctions and to study the pathogenesis of the disease, we have created transgenic mice expressing a truncated mouse cTnT allele analogous to one found in FHC patients. Mice expressing truncated cTnT at low (< 5%) levels develop cardiomyopathy and their hearts are significantly smaller (18-27%) than wild type. These animals also exhibit significant diastolic dysfunction and milder systolic dysfunction. Animals that express higher levels of transgene protein die within 24 h of birth. Transgenic mouse hearts demonstrate myocellular disarray and have a reduced number of cardiac myocytes that are smaller in size. These studies suggest that multiple cellular mechanisms result in the human disease, which is generally characterized by mild hypertrophy, but, also, frequent sudden death.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Heart/physiopathology , Mutation , Troponin/genetics , Animals , Base Sequence , Cardiomyopathy, Hypertrophic/physiopathology , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Troponin T
9.
Mol Cell Biol ; 10(6): 2625-37, 1990 Jun.
Article in English | MEDLINE | ID: mdl-1692957

ABSTRACT

The alpha 1-protease inhibitor (alpha 1-PI) proteins of mice are encoded by a group of genes whose members are expressed coordinately in a liver-abundant pattern and are regulated primarily at the transcriptional level. To better understand the developmental and tissue-specific regulation of this gene family, one member that is analogous to the human alpha 1-antitrypsin gene was chosen for study. Deletional analysis of the upstream regulatory region of this gene was performed, spanning from -10 kilobases to -80 base pairs relative to the transcriptional start site. Two functional positive cis-acting elements within the 522 bases immediately upstream of the start site for transcription were shown to modulate the level of expression from this promoter when introduced into human or mouse hepatoma cells, and a third region acted as a negative regulatory element in that its deletion resulted in a two- to sixfold increase of expression of a transfected minigene construct. Sequence comparison between the regulatory domains of two mouse alpha 1-PI genes and the human alpha 1-antitrypsin gene showed that the mouse gene contains a novel positive cis-acting element which is absent in human gene and that a specific eight-base-pair difference between species results in a strong positive cis-acting element in the human gene acting as a negative element in the mouse gene. An enhancer located approximately 3,000 base pairs upstream of the major start site for transcription was also identified. This element is position and orientation independent. Several different DNA-protein binding assays were used to demonstrate that each DNA segment with functional significance in transfection assays interacts specifically with proteins found in adult mouse liver nuclei. The major positive-acting element appeared to be specifically recognized by nuclear proteins found only in tissues that express alpha 1-PI, while the negative element binding proteins were ubiquitous. Thus, the distal regulatory domain including bases -3500 to -133 of this murine alpha 1-PI gene family member is more complex than was previously demonstrated. It is composed of a set of at least three additional functional cis-acting regulatory elements besides those which have been mapped by others and has a far upstream enhancer.


Subject(s)
Gene Expression Regulation , Genes , Transcription, Genetic , alpha 1-Antitrypsin/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Chromosome Deletion , Cloning, Molecular , Genes, Regulator , Humans , Mice , Molecular Sequence Data , Mutation , Oligonucleotide Probes , Plasmids , RNA/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Sequence Homology, Nucleic Acid , Transfection
11.
Genetics ; 118(2): 247-59, 1988 Feb.
Article in English | MEDLINE | ID: mdl-3129334

ABSTRACT

The 2B5 region on the X chromosome of Drosophila melanogaster forms an early ecdysone puff at the end of the third larval instar. The region contains a complex genetic locus, the Broad-Complex (BR-C) composed of four groups of fully complementing (br, rbp, l(1)2Bc, and l(1)2Bd) alleles, and classes of noncomplementing (npr 1) and partially noncomplementing l(1)2Bab alleles. BR-C mutants prevent metamorphosis, including the morphogenesis of imaginal discs. Results are presented that indicate that the BR-C contains two major functional domains. One, the br domain is primarily, if not exclusively, involved in the elongation and eversion of appendages by imaginal discs. The second, the l(1)2Bc domain, is primarily involved in the fusion of discs to form a continuous adult epidermis. Nonetheless, the two domains may encode products with related functions because in some situations mutants in both domains appear to affect similar developmental processes.


Subject(s)
Drosophila melanogaster/genetics , Mutation , Alleles , Animals , Crosses, Genetic , Drosophila melanogaster/growth & development , Female , Genes, Lethal , Heterozygote , Larva , Male , Phenotype , Pupa
SELECTION OF CITATIONS
SEARCH DETAIL
...