Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Neuro Oncol ; 17(12): 1599-608, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26116612

RÉSUMÉ

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas with minimal therapeutic opportunities. We observed that lipid droplets (LDs) accumulate in human MPNST cell lines and in primary human tumor samples. The goal of this study was to investigate the relevance of lipid metabolism to MPNST survival and as a possible therapeutic target. METHODS: Based on preliminary findings that MPNSTs accumulate LDs, we hypothesized that a deregulated lipid metabolism supports MPNST cell survival/proliferation rate. To test this, we examined respiration, role of fatty acid oxidation (FAO), and the enzyme fatty acid synthase involved in de novo fatty acid synthesis in MPNSTs using both genetic and pharmacological tools. RESULTS: We demonstrate that LDs accumulate in MPNST cell lines, primary human and mouse MPNST tumors, and neural crest cells. LDs from MPNST cells disappear on lipid deprivation, indicating that LDs can be oxidized as a source of energy. Inhibition of FAO decreased oxygen consumption and reduced MPNST survival, indicating that MPNST cells likely metabolize LDs through active FAO. FAO inhibition reduced oxygen consumption and survival even in the absence of exogenous lipids, indicating that lipids synthesized de novo can also be oxidized. Consequently, inhibition of de novo fatty acid synthesis, which is overexpressed in human MPNST cell lines, effectively reduced MPNST survival and delayed induction of tumor growth in vivo. CONCLUSION: Our results show that MPNSTs depend on lipid metabolic pathways and suggest that disrupting lipid metabolism could be a potential new strategy for the development of MPNST therapeutics.


Sujet(s)
Fatty acid synthases/métabolisme , Gouttelettes lipidiques/métabolisme , Neurinome/métabolisme , Cellules de Schwann/métabolisme , 4-Butyrolactone/analogues et dérivés , 4-Butyrolactone/pharmacologie , Animaux , Processus de croissance cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Fatty acid synthases/antagonistes et inhibiteurs , Humains , Souris , Souris de lignée C57BL , Crête neurale/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Proc Natl Acad Sci U S A ; 106(27): 11270-5, 2009 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-19541608

RÉSUMÉ

Mutations within the protein tyrosine phosphatase, SHP2, which is encoded by PTPN11, cause a significant proportion of Noonan syndrome (NS) cases, typically presenting with both cardiac disease and craniofacial abnormalities. Neural crest cells (NCCs) participate in both heart and skull formation, but the role of SHP2 signaling in NCC has not yet been determined. To gain insight into the role of SHP2 in NCC function, we ablated PTPN11 specifically in premigratory NCCs. SHP2-deficient NCCs initially exhibited normal migratory and proliferative patterns, but in the developing heart failed to migrate into the developing outflow tract. The embryos displayed persistent truncus arteriosus and abnormalities of the great vessels. The craniofacial deficits were even more pronounced, with large portions of the face and cranium affected, including the mandible and frontal and nasal bones. The data show that SHP2 activity in the NCC is essential for normal migration and differentiation into the diverse lineages found in the heart and skull and demonstrate the importance of NCC-based normal SHP2 activity in both heart and skull development, providing insight into the syndromic presentation characteristic of NS.


Sujet(s)
Coeur/embryologie , Crête neurale/embryologie , Crête neurale/enzymologie , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Crâne/embryologie , Crâne/enzymologie , Animaux , Différenciation cellulaire , Mouvement cellulaire , Malformations crâniofaciales/enzymologie , Malformations crâniofaciales/anatomopathologie , Régulation négative/génétique , Embryon de mammifère/enzymologie , Embryon de mammifère/anatomopathologie , Activation enzymatique , Extracellular Signal-Regulated MAP Kinases/métabolisme , Délétion de gène , Cardiopathies congénitales/enzymologie , Cardiopathies congénitales/anatomopathologie , Souris , Souris knockout , Crête neurale/cytologie , Phénotype , Phosphorylation
3.
Proc Natl Acad Sci U S A ; 105(48): 18930-5, 2008 Dec 02.
Article de Anglais | MEDLINE | ID: mdl-19017799

RÉSUMÉ

Noonan syndrome (NS) is the most common nonchromosomal genetic disorder associated with cardiovascular malformations. The most prominent cardiac defects in NS are pulmonary valve stenosis and hypertrophic cardiomyopathy. Gain-of-function mutations in the protein tyrosine phosphatase Shp2 have been identified in 50% of NS families. We created a NS mouse model with selective overexpression of mutant Shp2 (Q79R-Shp2) in the developing endocardial cushions. In our model, Cre recombinase driven by the Tie2 promoter irreversibly activates transgenic Q79R-Shp2 expression in the endothelial-derived cell lineage. Q79R-Shp2 expression resulted in embryonic lethality by embryonic day 14.5. Importantly, mutant embryos showed significantly enlarged endocardial cushions in the atrioventricular canal and in the outflow tract. In contrast, overexpression of wild-type Shp2 protein at comparable levels did not enhance endocardial cushion growth or alter the morphology of the mature adult valves. Expression of Q79R-Shp2 was accompanied by increased ERK1/2 activation in a subset of cells within the cushion mesenchyme, suggesting that hyperactivation of this signaling pathway may play a pathogenic role. To test this hypothesis in vivo, Q79R-Shp2-expressing mice were crossed with mice carrying either a homozygous ERK1 or a heterozygous ERK2 deletion. Deletion of ERK1 completely rescued the endocardial cushion phenotype, whereas ERK2 protein reduction did not affect endocardial cushion size. Constitutive hyperactivation of ERK1/2 signaling alone with a transgenic approach resulted in a phenocopy of the valvular phenotype. The data demonstrate both necessity and sufficiency of increased ERK activation downstream of Shp2 in mediating abnormal valve development in a NS mouse model.


Sujet(s)
Malformations/métabolisme , Valves cardiaques/malformations , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Syndrome de Noonan , Animaux , Malformations/génétique , Modèles animaux de maladie humaine , Activation enzymatique , Délétion de gène , Génotype , Coeur/anatomie et histologie , Coeur/embryologie , Valves cardiaques/métabolisme , Humains , Souris , Souris transgéniques , Mitogen-Activated Protein Kinase 1/génétique , Mitogen-Activated Protein Kinase 3/génétique , Syndrome de Noonan/génétique , Syndrome de Noonan/métabolisme , Phénotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Transduction du signal/physiologie
4.
J Mol Histol ; 38(6): 517-25, 2007 Dec.
Article de Anglais | MEDLINE | ID: mdl-17624499

RÉSUMÉ

Interneurons in the glomerular layer (GL) of the olfactory bulb represent a diverse set of cells, which can be identified by distinct expression of different neurotransmitters as well as calcium binding proteins. Using genetic based fate mapping, we show here that at least three of these different interneurons subtypes (i.e. dopaminergic, calbindin- and calretinin-expressing) derive from cells that express the homeobox genes Dlx5/6. The transcription factors ER81, Meis2, Pax6 and Sp8 have all been implicated in olfactory bulb interneuron development and each of these can be observed in Dlx5/6-derived periglomerular cells. Conversely, the T-box factors Tbr1 and Tbx21, which mark olfactory bulb projection neurons, are not expressed in the Dlx5/6-derived periglomerular cells. While the interneuron subtypes that are marked by Pax6 and Sp8 have been described, little information exists as to the specific subtypes that express ER81 or Meis2. We show here that ER81 is expressed in dopaminergic cells and in a subset of calretinin-expressing cells in the GL. Meis2 is found in dopaminergic and calbindin-expressing cells as well as in a subpopulation of the calretinin-expressing interneurons of the glomerular layer. These findings suggest that distinct transcriptional codes may underlie the differentiation of specific olfactory bulb interneuron subtypes.


Sujet(s)
Interneurones/métabolisme , Bulbe olfactif/cytologie , Bulbe olfactif/métabolisme , Facteurs de transcription/métabolisme , Animaux , Calbindine-2 , Calbindines , Protéines de liaison à l'ADN/métabolisme , Dopamine/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Interneurones/cytologie , Souris , Souris transgéniques , Protéine G liant le calcium S100/métabolisme , Cellules souches/métabolisme , Transcription génétique
5.
Cereb Cortex ; 17(11): 2585-92, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17259645

RÉSUMÉ

The standard method of detecting neurogenesis uses bromodeoxyuridine (BrdU) to label DNA synthesis followed by double labeling with neuronal markers. However, DNA synthesis may occur in events unrelated to neurogenesis including aneuploidy and abortive cell cycle reentry. Hence, it is important to confirm neurogenesis with methods other than BrdU incorporation. To this end, we have generated transgenic nestin-CreER mice that express tamoxifen-inducible Cre recombinase under the control of a nestin enhancer. When crossed with a ubiquitous Enhanced Green Fluorescent Protein (EGFP)-Cre-reporter line, the bitransgenic animals can reveal the nestin-positive progenitors and their progeny with EGFP after tamoxifen induction. This system has many applications including visualization of embryonic neural progenitors, detection of postnatally transformed radial glial cells, and labeling adult neural progenitors in the subventricular zone (SVZ). To examine the contribution of SVZ progenitors to cell replacement after stroke, tamoxifen-induced mice were challenged with focal ischemia or combined ischemia-hypoxia followed by BrdU injection. This analysis revealed only very few EGFP-positive cells outside the SVZ after focal ischemia but robust DNA synthesis by hippocampal neurons without immediate cell death following ischemia-hypoxia. These results suggest that the nestin-CreER system is a useful tool for detecting embryonic and adult neurogensis. They also confirm the existence of nonproliferative DNA synthesis by old neurons after experimental brain injury.


Sujet(s)
ADN/biosynthèse , Hypoxie-ischémie du cerveau/métabolisme , Protéines de filaments intermédiaires/métabolisme , Protéines de tissu nerveux/métabolisme , Neurones/métabolisme , Récepteurs des oestrogènes/métabolisme , Animaux , Apoptose/génétique , Protéines de filaments intermédiaires/génétique , Souris , Souris de lignée C57BL , Souris transgéniques , Protéines de tissu nerveux/génétique , Nestine , Récepteurs des oestrogènes/génétique
6.
Circ Res ; 98(12): 1547-54, 2006 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-16709902

RÉSUMÉ

Multipotent neural crest cells (NCCs) are a major extracardiac component of cardiovascular development. Although recognized as contributing cells to the arterial valves at early developmental stages, NCC persistence in the valves at later times or in the adult heart is controversial. We analyzed NCC persistence and contributions to both semilunar and atrioventricular (AV) valves in the mature heart. Two NCC-specific promoters driving Cre recombinase, Wnt1-Cre and P0-Cre, were mated with floxed reporter mice, R26R or CAG-CAT-EGFP, to map NCC fate. Hearts were analyzed before aorticopulmonary (AP) septation through adult stages. As previously demonstrated, strong NCC labeling was detected in ventral and dorsal outflow cushions before AP septation. In contrast to previous reports, we found that substantial numbers of labeled cells persisted in the semilunar valves in late fetal, neonatal, and adult hearts. Furthermore, NCCs were also found in the AV valves, almost exclusively in the septal leaflets. NCCs in the AV valves expressed melanocytic and neurogenic markers. However, cells labeled in the proximal cardiac conduction system exhibited neurogenic and gliagenic markers, whereas some NCCs expressed no differentiation specific markers. These results suggest that cardiac NCCs contribute to the mature valves and the cardiac conduction system and retain multipotent characteristics late in development.


Sujet(s)
Différenciation cellulaire , Système de conduction du coeur/cytologie , Valves cardiaques/embryologie , Valves cardiaques/croissance et développement , Crête neurale/cytologie , Vieillissement , Animaux , Animaux nouveau-nés , Aorte thoracique/cytologie , Marqueurs biologiques/métabolisme , Lignage cellulaire , Développement embryonnaire , Protéines à fluorescence verte , Système de conduction du coeur/embryologie , Système de conduction du coeur/croissance et développement , Valves cardiaques/cytologie , Souris , Souris transgéniques , Système nerveux/embryologie , Système nerveux/croissance et développement , Crête neurale/métabolisme
7.
Dev Growth Differ ; 47(3): 119-30, 2005 Apr.
Article de Anglais | MEDLINE | ID: mdl-15839997

RÉSUMÉ

Retinoic acid (RA), the active metabolite of vitamin A, regulates cellular growth and differentiation during embryonic development. In excess, this vitamin is also highly teratogenic to animals and humans. The neural crest is particularly sensitive to RA, and high levels adversely affect migration, proliferation and cell death. We investigated potential gene targets of RA associated with neural crest proliferation by determining RA-mediated changes in gene expression over time, using microarrays. Statistical analysis of the top ranked RA-regulated genes identified modest changes in multiple genes previously associated with cell cycle control and proliferation including the cyclin-dependent kinase inhibitors Cdkn1a (p21), Cdkn2b (p15(INK4b)), and Gas3/PMP22. The expression of p21 and p15(INK4b) contribute to decreased proliferation by blocking cell cycle progression at G1-S. This checkpoint is pivotal to decisions regulating proliferation, apoptosis, or differentiation. We have also confirmed the overexpression of Gas3/PMP22 in RA-treated neural crests, which is associated with cytoskeletal changes and increased apoptosis. Our results suggest that increases in multiple components of diverse regulatory pathways have an overall cumulative effect on cellular decisions. This heterogeneity contributes to the pleiotropic effects of RA, specifically those affecting proliferation and cell death.


Sujet(s)
Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Régulation de l'expression des gènes au cours du développement/effets des médicaments et des substances chimiques , Crête neurale/cytologie , Trétinoïne/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/métabolisme , Inhibiteur p15 de kinase cycline-dépendante , Inhibiteur p21 de kinase cycline-dépendante , Immunohistochimie , Souris , Souris de lignée ICR , Analyse sur microréseau , Protéines de la myéline/métabolisme , Crête neurale/effets des médicaments et des substances chimiques , Protéines suppresseurs de tumeurs/métabolisme
8.
Physiol Genomics ; 19(2): 184-97, 2004 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-15466718

RÉSUMÉ

Although retinoic acid (RA), the active form of vitamin A, is required for normal embryonic growth and development, it is also a powerful teratogen. Infants born to mothers exposed to retinoids during pregnancy have a 25-fold increased risk for malformations, nearly exclusively of cranial neural crest-derived tissues. To characterize neural crest cell responses to RA, we exposed murine crest cultures to teratogenic levels of RA and subjected their RNA to microarray-based gene expression profile analysis using Affymetrix MG-U74Av2 GeneChips. RNAs were isolated from independent cultures treated with 10(-6) M RA for 6, 12, 24, or 48 h. Statistical analyses of gene expression profile data facilitated identification of the 205 top-ranked differentially regulated genes whose expression was reproducibly changed by RA over time. Cluster analyses of these genes across the independently treated sample series revealed distinctive kinetic patterns of altered gene expression. The largest group was transiently affected within the first 6 h of exposure, representing early responding genes. Group 2 showed sustained induction by RA over all times, whereas group 3 was characterized by the suppression of a time-dependent expression increase normally seen in untreated cells. Additional patterns demonstrated time-dependent increased or decreased expression among genes not normally regulated to a significant extent. Gene function analysis revealed that more than one-third of all RA-regulated genes were associated with developmental regulation, including both canonical and noncanonical Wnt signaling pathways. Multiple genes associated with cell adhesion and cell cycle regulation, recognized targets for the biological effects of RA, were also affected. Taken together, these results support the hypothesis that the teratogenic effects of RA derive from reprogramming gene expression of a host of genes, which play critical roles during embryonic development regulating pathways that determine subsequent differentiation of cranial neural crest cells.


Sujet(s)
Régulation de l'expression des gènes au cours du développement/effets des médicaments et des substances chimiques , Crête neurale/composition chimique , Crête neurale/métabolisme , Trétinoïne/pharmacologie , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Adhérence cellulaire/génétique , Adhérence cellulaire/physiologie , Cellules cultivées , Lésions traumatiques des nerfs crâniens/induit chimiquement , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Analyse de profil d'expression de gènes/méthodes , Analyse de profil d'expression de gènes/statistiques et données numériques , Gènes/effets des médicaments et des substances chimiques , Gènes/physiologie , Gènes précoces/effets des médicaments et des substances chimiques , Gènes précoces/génétique , Souris , Crête neurale/cytologie , Crête neurale/effets des médicaments et des substances chimiques , Neurones/métabolisme , Séquençage par oligonucléotides en batterie/méthodes , Séquençage par oligonucléotides en batterie/statistiques et données numériques , Suppression génétique/effets des médicaments et des substances chimiques , Facteurs temps , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique
9.
Endocrinology ; 144(3): 1053-61, 2003 Mar.
Article de Anglais | MEDLINE | ID: mdl-12586782

RÉSUMÉ

PTHrP is a key developmental regulatory protein and a potent vasoactive agent. Previous studies have shown that mice lacking either the Pthrp or the PTH type 1 receptor (Pth1r) gene exhibit severe chondrodysplasia. In addition, in most genetic backgrounds, the receptor null mice die prenatally at midgestation, but the cause of death remains elusive. Here we show the loss of the Pth1r gene in C57BL6 mice leads to massive, abrupt cardiomyocyte death and embryonic lethality between embryonic days (E) E11.5 and E12.5. PTH1R mRNA was abundantly expressed in the developing wild-type mouse heart and cardiomyocytes from E11.5 embryos demonstrated acute increases in cAMP and increased Ca(2+)oscillations in response to PTHrP-(1-34)NH(2). Analyses of more than 300 embryos (E8-E14.5) from C57BL6/PTH1R +/- matings showed that PTH1R-/- mice survived until E11 with no obvious defects in any tissue. By E12, only 10% of the PTH1R-/- embryos survived and all PTH1R null mice were dead by E13. Ultrastructural and histological analysis revealed striking mitochondrial abnormalities at E11.5 and precipitous cardiomyocyte death between E12.0 and E12.5, followed by degenerative changes in the liver and massive necrosis of other tissues. No abnormalities were observed in the yolk sac or placenta implicating the heart degeneration as the primary cause of death. Taken together, these findings indicate that the PTH1R is required for the development of normal cardiomyocyte function.


Sujet(s)
Mort cellulaire , Mort foetale/étiologie , Âge gestationnel , Myocarde/ultrastructure , Récepteurs à l'hormone parathyroïdienne/déficit , Animaux , Apoptose , Femelle , Coeur/embryologie , Humains , Hybridation in situ , Méthode TUNEL , Souris , Souris de lignée C57BL , Souris knockout , Microscopie électronique , Myocarde/composition chimique , Grossesse , ARN messager/analyse , Récepteur de la parathormone de type 1 , Récepteurs à l'hormone parathyroïdienne/génétique , Récepteurs à l'hormone parathyroïdienne/physiologie , Transduction du signal
10.
J Mol Cell Cardiol ; 34(10): 1411-23, 2002 Oct.
Article de Anglais | MEDLINE | ID: mdl-12393001

RÉSUMÉ

BACKGROUND: Beta-MHC-hRARalpha transgenic mice express a constitutively active (truncated) form of the human retinoic acid receptor which triggers development of dilated cardiomyopathy. In those hearts, we studied expression of gap junction proteins in relation to electrical impulse propagation. METHODS AND RESULTS: As compared to wildtype mice, hearts of 4-6 month old mice with 7-12 inserted hRARalpha copies are marked by an increased heart weight/body weight- and heart weight/tibia length ratio. 3-extremity lead ECGs revealed prolongation of the Q-j interval suggesting delayed ventricular activation. Mapping of electrical activity of epi- and endocardial left ventricular free wall revealed activation delay, increased heterogeneity in conduction and regional conduction block. Ventricular tachycardias did not occur spontaneously nor could be induced by ventricular pacing. Immunohistochemical analysis showed profound and heterogeneous redistribution and down-regulation of the gap junction protein connexin43 (Cx43) in the left ventricular free wall. Here, hRARalpha expression induced re-expression of the hypertrophic markers alpha-skeletal actin and beta-MHC, and in 3 out of 10 severely affected mice, re-expression of Cx40. Concomitant with changes in expression/distribution of Cx43, changes in expression and distribution of beta-catenin and N-cadherin (two other intercalated disk associated proteins) were observed. CONCLUSIONS: Beta-MHC-hRARalpha transgenic hearts show heterogeneous re-expression of (early) sarcomeric genes while expression of connexin43, N-cadherin and beta-catenin is down-regulated. We postulate that the resulting aberrant ventricular activation does not trigger development of lethal arrhythmias due to the small size of remaining healthy ventricular tissue where the transgene is not expressed.


Sujet(s)
Cardiomégalie/anatomopathologie , Jonctions communicantes/métabolisme , Myocarde/anatomopathologie , Transduction du signal , Trétinoïne/métabolisme , Actines/métabolisme , Animaux , Marqueurs biologiques/analyse , Technique de Western , Poids , Cadhérines/métabolisme , Connexine 43/métabolisme , Protéines du cytosquelette/métabolisme , Régulation négative , Stimulation électrique , Électrocardiographie , Souris , Souris transgéniques , Taille d'organe , Récepteurs à l'acide rétinoïque/génétique , Récepteurs à l'acide rétinoïque/métabolisme , Transactivateurs/métabolisme , bêta-Caténine
11.
Cardiovasc Toxicol ; 2(1): 25-39, 2002.
Article de Anglais | MEDLINE | ID: mdl-12189278

RÉSUMÉ

Vitamin A and related retinoids are critical regulators of normal cardiovascular development. Extreme variations in retinoid levels, too little or too much, dramatically alter embryonic morphogenesis that has teratogenic consequences, including effects on the heart and great vessels. Specific cardiovascular targets of retinoid action include effects on the anteroposterior patterning of the early heart, left-right decisions and cardiac situs, endocardial cushion formation, and, in particular, the neural crest. The cardiovascular defects produced are remarkably similar in deficiency and excess, suggesting modulation of common developmental or cellular processes by different levels of retinoids. The isolation of nuclear receptors that mediate retinoid action has led to the identification of some genes directly involved in the regulation of these processes and other gene products that may be affected more indirectly. This review will examine the mechanism of retinoid action, the requirements for vitamin A during normal heart development, and the consequences of nonphysiologic or teratogenic exposure.


Sujet(s)
Malformations cardiovasculaires/induit chimiquement , Malformations cardiovasculaires/anatomopathologie , Rétinoïdes/physiologie , Rétinoïdes/toxicité , Animaux , Système cardiovasculaire/croissance et développement , Humains
12.
Nat Med ; 8(7): 725-30, 2002 Jul.
Article de Anglais | MEDLINE | ID: mdl-12053174

RÉSUMÉ

Loss of cardiomyocytes through programmed cell death is a key event in the development of heart failure, but the inciting molecular mechanisms are largely unknown. We used microarray analysis to identify a genetic program for myocardial apoptosis in Gq-mediated and pressure-overload cardiac hypertrophy. A critical component of this apoptotic program was Nix/Bnip3L. Nix localized to mitochondria and caused release of cytochrome c, activation of caspase-3 and apoptotic cell death, when expressed in HEK293 fibroblasts. A previously undescribed truncated Nix isoform, termed sNix, was not targeted to mitochondria but heterodimerized with Nix and protected against Nix-mediated apoptosis. Forced in vivo myocardial expression of Nix resulted in apoptotic cardiomyopathy and rapid death. Conversely, sNix protected against apoptotic peripartum cardiomyopathy in G(alpha)q-overexpressors. Thus, Nix/Bnip3L is upregulated in myocardial hypertrophy, and is both necessary and sufficient for Gq-mediated apoptosis of cardiomyocytes and resulting hypertrophy decompensation.


Sujet(s)
Apoptose/physiologie , Cardiomyopathie hypertrophique/anatomopathologie , Protéines membranaires/physiologie , Mitochondries/physiologie , Protéines proto-oncogènes , Protéines suppresseurs de tumeurs , Animaux , Clonage moléculaire , Humains , Protéines membranaires/génétique , Souris , Souris transgéniques , Mitochondries/anatomopathologie , Données de séquences moléculaires , Protéines recombinantes/métabolisme , RT-PCR
13.
Cardiovasc Res ; 54(1): 105-16, 2002 Apr.
Article de Anglais | MEDLINE | ID: mdl-12062367

RÉSUMÉ

OBJECTIVE: Impaired myocyte Ca2+ handling is a common characteristic of failing hearts and increases in calcineurin activity, a Ca2+-sensitive phosphatase, have been implicated in heart failure phenotype. Transgenic mice with cardiac-specific expression of an active form of calcineurin display depressed function, hypertrophy and heart failure. We examined whether defects in cardiomyocyte Ca2+ handling properties contribute to the impaired cardiac function in calcineurin transgenic mice. METHODS: The levels of SR Ca2+ handling proteins, SR Ca2+ transport function and cardiomyocyte mechanics, as well as Ca2+ kinetics were examined in mice overexpressing a constitutively active form of calcineurin. RESULTS: Transgenic expression of activated calcineurin catalytic subunit resulted in significant protein increases (66%) in SERCA2 and decreases (35%) in phospholamban, as well as enhanced (approximately 80%) phospholamban phosphorylation. These alterations in the SR Ca2+-transport proteins resulted in increased V(max) and Ca2+-affinity of SERCA2. The myofibrillar Mg-ATPase activity was also significantly increased at pCa>6.0. The enhanced SR Ca2+ handling and Mg-ATPase activity reflected significant elevation in myocyte contractile parameters (3-fold), Ca2+ transient amplitude (1.5-fold) and the rate of Ca2+ signal decay (2-fold). In contrast, in vivo cardiac function assessed by echocardiography, indicated severely depressed contractility in calcineurin hearts. The apparent disparity in contractile properties between the cellular and multicellular preparations may be partially due to tissue remodeling, including interstitial fibrosis and a marked reduction (45%), dephosphorylation (81%) and redistribution of the gap junctional protein connexin-43, which could compromise intercellular communication. CONCLUSION: Despite enhanced SR Ca2+ handling and contractility in myocytes, pathological remodeling and defects in intercellular coupling may underlie contractile dysfunction of the calcineurin hearts.


Sujet(s)
Calcineurine/génétique , Calcium/métabolisme , Défaillance cardiaque/physiopathologie , Contraction myocardique , Animaux , Ca(2+) Mg(2+)-ATPase/métabolisme , Calcineurine/métabolisme , Protéines de liaison au calcium/métabolisme , Cellules cultivées , Connexine 43/métabolisme , Électrocardiographie , Défaillance cardiaque/métabolisme , Immunotransfert/méthodes , Souris , Souris transgéniques , Myofibrilles/enzymologie , Réticulum sarcoplasmique/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...