Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Circ Res ; 134(8): 1006-1022, 2024 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-38506047

RÉSUMÉ

BACKGROUND: In heart failure, signaling downstream the ß2-adrenergic receptor is critical. Sympathetic stimulation of ß2-adrenergic receptor alters cAMP (cyclic adenosine 3',5'-monophosphate) and triggers PKA (protein kinase A)-dependent phosphorylation of proteins that regulate cardiac function. cAMP levels are regulated in part by PDEs (phosphodiesterases). Several AKAPs (A kinase anchoring proteins) regulate cardiac function and are proposed as targets for precise pharmacology. AKAP12 is expressed in the heart and has been reported to directly bind ß2-adrenergic receptor, PKA, and PDE4D. However, its roles in cardiac function are unclear. METHODS: cAMP accumulation in real time downstream of the ß2-adrenergic receptor was detected for 60 minutes in live cells using the luciferase-based biosensor (GloSensor) in AC16 human-derived cardiomyocyte cell lines overexpressing AKAP12 versus controls. Cardiomyocyte intracellular calcium and contractility were studied in adult primary cardiomyocytes from male and female mice overexpressing cardiac AKAP12 (AKAP12OX) and wild-type littermates post acute treatment with 100-nM isoproterenol (ISO). Systolic cardiac function was assessed in mice after 14 days of subcutaneous ISO administration (60 mg/kg per day). AKAP12 gene and protein expression levels were evaluated in left ventricular samples from patients with end-stage heart failure. RESULTS: AKAP12 upregulation significantly reduced total intracellular cAMP levels in AC16 cells through PDE8. Adult primary cardiomyocytes from AKAP12OX mice had significantly reduced contractility and impaired calcium handling in response to ISO, which was reversed in the presence of the selective PDE8 inhibitor (PF-04957325). AKAP12OX mice had deteriorated systolic cardiac function and enlarged left ventricles. Patients with end-stage heart failure had upregulated gene and protein levels of AKAP12. CONCLUSIONS: AKAP12 upregulation in cardiac tissue is associated with accelerated cardiac dysfunction through the AKAP12-PDE8 axis.


Sujet(s)
3',5'-Cyclic-AMP Phosphodiesterases , Cardiopathies , Récepteurs adrénergiques , Animaux , Femelle , Humains , Mâle , Souris , 3',5'-Cyclic-AMP Phosphodiesterases/génétique , 3',5'-Cyclic-AMP Phosphodiesterases/métabolisme , Protéines d'ancrage aux protéines kinases A/génétique , Protéines d'ancrage aux protéines kinases A/métabolisme , Calcium/métabolisme , Protéines du cycle cellulaire/génétique , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cardiopathies/métabolisme , Défaillance cardiaque/génétique , Défaillance cardiaque/métabolisme , Isoprénaline/pharmacologie , Myocytes cardiaques/métabolisme , Récepteurs adrénergiques/métabolisme , Régulation positive
2.
Endocrinology ; 163(3)2022 03 01.
Article de Anglais | MEDLINE | ID: mdl-35134138

RÉSUMÉ

Female mice homozygous for an engineered Gnrhr E90K mutation have reduced gonadotropin-releasing hormone signaling, leading to infertility. Their ovaries have numerous antral follicles but no corpora lutea, indicating a block to ovulation. These mutants have high levels of circulating estradiol and low progesterone, indicating a state of persistent estrus. This mouse model provided a unique opportunity to examine the lack of cyclic levels of ovarian hormones on uterine gland biology. Although uterine gland development appeared similar to controls during prepubertal development, it was compromised during adolescence in the mutants. By age 20 weeks, uterine gland development was comparable to controls, but pathologies, including cribriform glandular structures, were observed. Induction of ovulations by periodic human chorionic gonadotropin treatment did not rescue postpubertal uterine gland development. Interestingly, progesterone receptor knockout mice, which lack progesterone signaling, also have defects in postpubertal uterine gland development. However, progesterone treatment did not rescue postpubertal uterine gland development. These studies indicate that chronically elevated levels of estradiol with low progesterone and therefore an absence of cyclic ovarian hormone secretion disrupts postpubertal uterine gland development and homeostasis.


Sujet(s)
Oestradiol/sang , Oestrus/physiologie , Infertilité féminine/génétique , Progestérone/sang , Récepteurs à la gonadolibérine/génétique , Utérus/croissance et développement , Animaux , Gonadotrophine chorionique/pharmacologie , Oestrus/effets des médicaments et des substances chimiques , Femelle , Infertilité féminine/sang , Souris , Souris knockout , Follicule ovarique/effets des médicaments et des substances chimiques , Ovulation/effets des médicaments et des substances chimiques , Progestérone/pharmacologie , Utérus/effets des médicaments et des substances chimiques
3.
J Cachexia Sarcopenia Muscle ; 12(6): 2174-2186, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34704401

RÉSUMÉ

BACKGROUND: Skeletal muscle atrophy is a debilitating complication of many chronic diseases, disuse conditions, and ageing. Genome-wide gene expression analyses have identified that elevated levels of microRNAs encoded by the H19X locus are among the most significant changes in skeletal muscles in a wide scope of human cachectic conditions. We have previously reported that the H19X locus is important for the establishment of striated muscle fate during embryogenesis. However, the role of H19X-encoded microRNAs in regulating skeletal mass in adults is unknown. METHODS: We have created a transgenic mouse strain in which ectopic expression of miR-322/miR-503 is driven by the skeletal muscle-specific muscle creatine kinase promoter. We also used an H19X mutant mouse strain in which transcription from the locus is interrupted by a gene trap. Animal phenotypes were analysed by standard histological methods. Underlying mechanisms were explored by using transcriptome profiling and validated in the two animal models and cultured myotubes. RESULTS: Our results demonstrate that the levels of H19X microRNAs are inversely related to postnatal skeletal muscle growth. Targeted overexpression of miR-322/miR-503 impeded skeletal muscle growth. The weight of gastrocnemius muscles of transgenic mice was only 54.5% of the counterparts of wild-type littermates. By contrast, interruption of transcription from the H19X locus stimulates postnatal muscle growth by 14.4-14.9% and attenuates the loss of skeletal muscle mass in response to starvation by 12.8-21.0%. Impeded muscle growth was not caused by impaired IGF1/AKT/mTOR signalling or a hyperactive ubiquitin-proteasome system, instead accompanied by markedly dropped abundance of translation initiation factors in transgenic mice. miR-322/miR-503 directly targets eIF4E, eIF4G1, eIF4B, eIF2B5, and eIF3M. CONCLUSIONS: Our study illustrates a novel pathway wherein H19X microRNAs regulate skeletal muscle growth and atrophy through regulating the abundance of translation initiation factors, thereby protein synthesis. The study highlights how translation initiation factors lie at the crux of multiple signalling pathways that control skeletal muscle mass.


Sujet(s)
microARN , Amyotrophie , Animaux , Souris , microARN/génétique , Fibres musculaires squelettiques , Muscles squelettiques/anatomopathologie , Amyotrophie/génétique , Amyotrophie/anatomopathologie , Facteurs initiation chaîne peptidique
4.
FASEB J ; 34(1): 555-570, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31914652

RÉSUMÉ

Muscle wasting, or muscle atrophy, can occur with age, injury, and disease; it affects the quality of life and complicates treatment. Insulin-like growth factor 1 (IGF1) is a key positive regulator of muscle mass. The IGF1/Igf1 gene encodes multiple protein isoforms that differ in tissue expression, potency, and function, particularly in cellular proliferation and differentiation, as well as in systemic versus localized signaling. Genome engineering is a novel strategy for increasing gene expression and has the potential to recapitulate the diverse biology seen in IGF1 signaling through the overexpression of multiple IGF1 isoforms. Using a CRISPR-Cas9 gene activation approach, we showed that the expression of multiple IGF1 or Igf1 mRNA variants can be increased in human and mouse skeletal muscle myoblast cell lines using a single-guide RNA (sgRNA). We found increased IGF1 protein levels in the cell culture media and increased cellular phosphorylation of AKT1, the main effector of IGF1 signaling. We also showed that the expression of Class 1 or Class 2 mRNA variants can be selectively increased by changing the sgRNA target location. The expression of multiple IGF1 or Igf1 mRNA transcript variants in human and mouse skeletal muscle myoblasts promoted myotube differentiation and prevented dexamethasone-induced atrophy in myotubes in vitro. Our findings suggest that this novel approach for enhancing IGF1 signaling has potential therapeutic applications for treating skeletal muscle atrophy.


Sujet(s)
Systèmes CRISPR-Cas , Différenciation cellulaire , Facteur de croissance IGF-I/métabolisme , Muscles squelettiques/cytologie , Amyotrophie/anatomopathologie , Myoblastes/cytologie , Activation de la transcription , Animaux , Anti-inflammatoires/pharmacologie , Séquence nucléotidique , Prolifération cellulaire , Cellules cultivées , Dexaméthasone/pharmacologie , Humains , Facteur de croissance IGF-I/génétique , Souris , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Amyotrophie/induit chimiquement , Amyotrophie/métabolisme , Myoblastes/effets des médicaments et des substances chimiques , Myoblastes/métabolisme , Phosphorylation , Isoformes de protéines , ARN messager/génétique , ARN messager/métabolisme , Similitude de séquences , Transduction du signal
5.
Fundam Clin Pharmacol ; 33(1): 25-30, 2019 Feb.
Article de Anglais | MEDLINE | ID: mdl-29959870

RÉSUMÉ

Inbred mouse strains are the most widely used mammalian model organism in biomedical research owing to ease of genetic manipulation and short lifespan; however, each inbred strain possesses a unique repertoire of deleterious homozygous alleles that can make a specific strain more susceptible to a particular disease. In the current study, we report dystrophic cardiac calcinosis (DCC) in C.B-17 SCID male mice at 10 weeks of age with no significant change in cardiac function. Acquisition of DCC was characterized by myocardial injury, fibrosis, calcification, and necrosis of the tissue. At 10 weeks of age, 38% of the C.B-17 SCID mice from two different commercial colonies exhibited significant calcinosis on the ventricular epicardium, predominantly on the right ventricle. The frequency of calcinosis was more than 50% for mice obtained from Taconic's Cambridge City colony and 25% for mice obtained from Taconic's German Town colony. Interestingly, the DCC phenotype did not affect cardiac function at 10 weeks of age. No differences in echocardiography or electrocardiography were observed between the calcinotic and non-calcinotic mice from either colony. Our findings suggest that C.B-17 SCID mice exhibit DCC as early as 10 weeks of age with no significant impact on cardiac function. This strain of mice should be cautiously considered for the study of cardiac physiology.


Sujet(s)
Calcinose/anatomopathologie , Cardiomyopathies/physiopathologie , Modèles animaux de maladie humaine , Péricarde/anatomopathologie , Animaux , Échocardiographie/méthodes , Électrocardiographie/méthodes , Mâle , Souris , Lignées consanguines de souris , Souris SCID , Phénotype
6.
Methods Mol Biol ; 1668: 167-176, 2017.
Article de Anglais | MEDLINE | ID: mdl-28842909

RÉSUMÉ

Mammalian myogenesis occurs in two distinct phases, primary and secondary, which are temporally separated. The primary wave occurs shortly after somitogenesis producing embryonic myofibers. The secondary wave occurs after somitogenesis producing fetal myofibers that form adjacent to the embryonic myofibers. The myogenic cells that give rise to these two waves have distinct characteristics as do the myofibers they produce. The objective of this chapter is to describe our methods for quantifying embryonic and fetal myofiber development in mouse embryos using immunofluorescence.


Sujet(s)
Embryon de mammifère/embryologie , Foetus/embryologie , Technique d'immunofluorescence/méthodes , Développement musculaire/physiologie , Fibres musculaires squelettiques/physiologie , Muscles squelettiques/embryologie , Animaux , Prolifération cellulaire , Femelle , Colorants fluorescents/composition chimique , Souris , Souris knockout , Myoblastes/métabolisme , Imagerie optique , Somites/métabolisme
7.
J Cell Sci ; 130(15): 2551-2563, 2017 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-28600325

RÉSUMÉ

Nucleosome assembly proceeds through DNA replication-coupled or replication-independent mechanisms. For skeletal myocytes, whose nuclei have permanently exited the cell cycle, replication-independent assembly is the only mode available for chromatin remodeling. For this reason, any nucleosome composition alterations accompanying transcriptional responses to physiological signals must occur through a DNA replication-independent pathway. HIRA is the histone chaperone primarily responsible for replication-independent incorporation of histone variant H3.3 across gene bodies and regulatory regions. Thus, HIRA would be expected to play an important role in epigenetically regulating myocyte gene expression. The objective of this study was to determine the consequence of eliminating HIRA from mouse skeletal myocytes. At 6 weeks of age, myofibers lacking HIRA showed no pathological abnormalities; however, genes involved in transcriptional regulation were downregulated. By 6 months of age, myofibers lacking HIRA exhibited hypertrophy, sarcolemmal perforation and oxidative damage. Genes involved in muscle growth and development were upregulated, but those associated with responses to cellular stresses were downregulated. These data suggest that elimination of HIRA produces a hypertrophic response in skeletal muscle and leaves myofibers susceptible to stress-induced degeneration.


Sujet(s)
Protéines du cycle cellulaire/déficit , Chaperons d'histones/déficit , Muscles squelettiques/métabolisme , Maladies musculaires/métabolisme , Stress oxydatif , Facteurs de transcription/déficit , Animaux , Hypertrophie , Souris , Souris transgéniques , Muscles squelettiques/anatomopathologie , Maladies musculaires/génétique , Maladies musculaires/anatomopathologie
8.
Oncotarget ; 8(27): 43866-43877, 2017 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-28410206

RÉSUMÉ

Cancer is genetically heterogeneous regarding to molecular genetic characteristics and pathogenic pathways. A wide spectrum of biomarkers, including DNA markers, is used in determining genomic instability, molecular subtype determination and disease prognosis, and estimating sensitivity to different drugs in clinical practice. In a previous study, we developed highly effective DNA markers using improved random amplified polymorphic DNA (RAPD) with high-GC primers, which is a valuable approach for the genetic authentication of medicinal plants. In this study, we applied this effective DNA marker technique to generate genetic fingerprints that detect genomic alterations in human breast cancer tissues and then developed sequence-characterized amplified region (SCAR) markers. Three SCAR markers (BC10-1, BC13-4 and BC31-2) had high levels of genomic DNA amplification in breast cancer. The PHKG2 and RNF40 genes are either overlapping or close to the sequences of SCAR marker BC13-4, while SCAR marker BC10-1 is in the intron and overlap the DPEP1 gene, suggesting that alterations in the expression of these genes could contribute to cancer progression. Screening of breast cancer cell lines showed that the mRNA expression levels for the PHKG2 and DPEP1 were lower in non-tumorigenic mammary epithelial cell MCF10A, but elevated in other cell lines. The DPEP1 mRNA level in invasive ductal carcinoma specimens was significantly higher than that of the adjacent normal tissues in women. Taken together, high-GC RAMP-PCR provides greater efficacy in measuring genomic DNA amplifications, deletion or copy number variations. Furthermore, SCAR markers BC10-1 and BC13-4 might be useful diagnostic markers for breast cancer carcinomas.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein/diagnostic , Tumeurs du sein/génétique , Amplification de gène , Génomique , Adulte , Sujet âgé , Composition en bases nucléiques , Séquence nucléotidique , Carcinome canalaire du sein/diagnostic , Carcinome canalaire du sein/génétique , Clonage moléculaire , Amorces ADN , Dipeptidases/génétique , Femelle , Protéines liées au GPI/génétique , Génomique/méthodes , Humains , Adulte d'âge moyen , Grading des tumeurs , Invasion tumorale , Stadification tumorale , ARN messager/génétique , Technique RAPD , Analyse de séquence d'ADN
9.
Sci Rep ; 6: 31457, 2016 08 19.
Article de Anglais | MEDLINE | ID: mdl-27538477

RÉSUMÉ

Mesp1 directs multipotential cardiovascular cell fates, even though it's transiently induced prior to the appearance of the cardiac progenitor program. Tracing Mesp1-expressing cells and their progeny allows isolation and characterization of the earliest cardiovascular progenitor cells. Studying the biology of Mesp1-CPCs in cell culture and ischemic disease models is an important initial step toward using them for heart disease treatment. Because of Mesp1's transitory nature, Mesp1-CPC lineages were traced by following EYFP expression in murine Mesp1(Cre/+); Rosa26(EYFP/+) ES cells. We captured EYFP+ cells that strongly expressed cardiac mesoderm markers and cardiac transcription factors, but not pluripotent or nascent mesoderm markers. BMP2/4 treatment led to the expansion of EYFP+ cells, while Wnt3a and Activin were marginally effective. BMP2/4 exposure readily led EYFP+ cells to endothelial and smooth muscle cells, but inhibition of the canonical Wnt signaling was required to enter the cardiomyocyte fate. Injected mouse pre-contractile Mesp1-EYFP+ CPCs improved the survivability of injured mice and restored the functional performance of infarcted hearts for at least 3 months. Mesp1-EYFP+ cells are bona fide CPCs and they integrated well in infarcted hearts and emerged de novo into terminally differentiated cardiac myocytes, smooth muscle and vascular endothelial cells.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Infarctus du myocarde/thérapie , Transplantation de cellules souches , Cellules souches/métabolisme , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Protéine morphogénétique osseuse de type 2/pharmacologie , Protéine morphogénétique osseuse de type 4/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignage cellulaire , Coeur/imagerie diagnostique , Mâle , Mésoderme/cytologie , Mésoderme/métabolisme , Souris , Souris SCID , Cellules souches embryonnaires de souris/cytologie , Cellules souches embryonnaires de souris/métabolisme , Infarctus du myocarde/anatomopathologie , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Séquençage par oligonucléotides en batterie , Transduction du signal/effets des médicaments et des substances chimiques , Cellules souches/cytologie , Transcriptome , Protéine Wnt3A/métabolisme
10.
Dis Model Mech ; 9(3): 335-45, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26935106

RÉSUMÉ

HIRA is the histone chaperone responsible for replication-independent incorporation of histone variant H3.3 within gene bodies and regulatory regions of actively transcribed genes, and within the bivalent promoter regions of developmentally regulated genes. The HIRA gene lies within the 22q11.2 deletion syndrome critical region; individuals with this syndrome have multiple congenital heart defects. Because terminally differentiated cardiomyocytes have exited the cell cycle, histone variants should be utilized for the bulk of chromatin remodeling. Thus, HIRA is likely to play an important role in epigenetically defining the cardiac gene expression program. In this study, we determined the consequence of HIRA deficiency in cardiomyocytes in vivo by studying the phenotype of cardiomyocyte-specific Hira conditional-knockout mice. Loss of HIRA did not perturb heart development, but instead resulted in cardiomyocyte hypertrophy and susceptibility to sarcolemmal damage. Cardiomyocyte degeneration gave way to focal replacement fibrosis and impaired cardiac function. Gene expression was widely altered in Hira conditional-knockout hearts. Significantly affected pathways included responses to cellular stress, DNA repair and transcription. Consistent with heart failure, fetal cardiac genes were re-expressed in the Hira conditional knockout. Our results suggest that transcriptional regulation by HIRA is crucial for cardiomyocyte homeostasis.


Sujet(s)
Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Protéines du cycle cellulaire/déficit , Chaperons d'histones/déficit , Myocytes cardiaques/métabolisme , Sarcolemme/métabolisme , Sarcolemme/anatomopathologie , Facteurs de transcription/déficit , Animaux , Apoptose/génétique , Cardiomégalie/génétique , Cardiomégalie/physiopathologie , Protéines du cycle cellulaire/métabolisme , Prolifération cellulaire , Altération de l'ADN/génétique , Réparation de l'ADN/génétique , Foetus/métabolisme , Régulation de l'expression des gènes , Tests de la fonction cardiaque , Chaperons d'histones/métabolisme , Souris knockout , Myocytes cardiaques/anatomopathologie , Spécificité d'organe , Stress oxydatif/génétique , Reproductibilité des résultats , Stress physiologique/génétique , Facteurs de transcription/métabolisme , Transcriptome/génétique
11.
Dis Model Mech ; 9(3): 347-59, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26935107

RÉSUMÉ

The Smyd1 gene encodes a lysine methyltransferase specifically expressed in striated muscle. Because Smyd1-null mouse embryos die from heart malformation prior to formation of skeletal muscle, we developed a Smyd1 conditional-knockout allele to determine the consequence of SMYD1 loss in mammalian skeletal muscle. Ablation of SMYD1 specifically in skeletal myocytes after myofiber differentiation using Myf6(cre) produced a non-degenerative myopathy. Mutant mice exhibited weakness, myofiber hypotrophy, prevalence of oxidative myofibers, reduction in triad numbers, regional myofibrillar disorganization/breakdown and a high percentage of myofibers with centralized nuclei. Notably, we found broad upregulation of muscle development genes in the absence of regenerating or degenerating myofibers. These data suggest that the afflicted fibers are in a continual state of repair in an attempt to restore damaged myofibrils. Disease severity was greater for males than females. Despite equivalent expression in all fiber types, loss of SMYD1 primarily affected fast-twitch muscle, illustrating fiber-type-specific functions for SMYD1. This work illustrates a crucial role for SMYD1 in skeletal muscle physiology and myofibril integrity.


Sujet(s)
Noyau de la cellule/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines du muscle/métabolisme , Amyotrophie/enzymologie , Myofibrilles/enzymologie , Myofibrilles/anatomopathologie , Facteurs de transcription/métabolisme , Animaux , Femelle , Mâle , Souris knockout , Développement musculaire/génétique , Fibres musculaires à contraction rapide/métabolisme , Fibres musculaires à contraction rapide/ultrastructure , Force musculaire , Amyotrophie/anatomopathologie , Myofibrilles/ultrastructure , Taille d'organe , Oxydoréduction , Régénération , Sarcolemme/métabolisme , Régulation positive/génétique
12.
Dev Biol ; 410(1): 86-97, 2016 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-26688546

RÉSUMÉ

The SMYD (SET and MYND domain) family of lysine methyltransferases harbor a unique structure in which the methyltransferase (SET) domain is intervened by a zinc finger protein-protein interaction MYND domain. SMYD proteins methylate both histone and non-histone substrates and participate in diverse biological processes including transcriptional regulation, DNA repair, proliferation and apoptosis. Smyd1 is unique among the five family members in that it is specifically expressed in striated muscles. Smyd1 is critical for development of the right ventricle in mice. In zebrafish, Smyd1 is necessary for sarcomerogenesis in fast-twitch muscles. Smyd1 is expressed in the skeletal muscle lineage throughout myogenesis and in mature myofibers, shuttling from nucleus to cytosol during myoblast differentiation. Because of this expression pattern, we hypothesized that Smyd1 plays multiple roles at different stages of myogenesis. To determine the role of Smyd1 in mammalian myogenesis, we conditionally eliminated Smyd1 from the skeletal muscle lineage at the myoblast stage using Myf5(cre). Deletion of Smyd1 impaired myoblast differentiation, resulted in fewer myofibers and decreased expression of muscle-specific genes. Muscular defects were temporally restricted to the second wave of myogenesis. Thus, in addition to the previously described functions for Smyd1 in heart development and skeletal muscle sarcomerogenesis, these results point to a novel role for Smyd1 in myoblast differentiation.


Sujet(s)
Protéines de liaison à l'ADN/physiologie , Développement musculaire , Protéines du muscle/physiologie , Facteurs de transcription/physiologie , Animaux , Différenciation cellulaire , Cellules cultivées , Protéines de liaison à l'ADN/analyse , Souris , Fibres musculaires squelettiques , Protéines du muscle/analyse , Myoblastes/cytologie , Facteurs de transcription/analyse
13.
PLoS One ; 10(3): e0121765, 2015.
Article de Anglais | MEDLINE | ID: mdl-25803368

RÉSUMÉ

Smyd1/Bop is an evolutionary conserved histone methyltransferase previously shown by conventional knockout to be critical for embryonic heart development. To further explore the mechanism(s) in a cell autonomous context, we conditionally ablated Smyd1 in the first and second heart fields of mice using a knock-in (KI) Nkx2.5-cre driver. Robust deletion of floxed-Smyd1 in cardiomyocytes and the outflow tract (OFT) resulted in embryonic lethality at E9.5, truncation of the OFT and right ventricle, and additional defects consistent with impaired expansion and proliferation of the second heart field (SHF). Using a transgenic (Tg) Nkx2.5-cre driver previously shown to not delete in the SHF and OFT, early embryonic lethality was bypassed and both ventricular chambers were formed; however, reduced cardiomyocyte proliferation and other heart defects resulted in later embryonic death at E11.5-12.5. Proliferative impairment prior to both early and mid-gestational lethality was accompanied by dysregulation of transcripts critical for endoplasmic reticulum (ER) stress. Mid-gestational death was also associated with impairment of oxidative stress defense-a phenotype highly similar to the previously characterized knockout of the Smyd1-interacting transcription factor, skNAC. We describe a potential feedback mechanism in which the stress response factor Tribbles3/TRB3, when directly methylated by Smyd1, acts as a co-repressor of Smyd1-mediated transcription. Our findings suggest that Smyd1 is required for maintaining cardiomyocyte proliferation at minimally two different embryonic heart developmental stages, and its loss leads to linked stress responses that signal ensuing lethality.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Stress du réticulum endoplasmique , Coeur/croissance et développement , Protéines du muscle/métabolisme , Myocarde/cytologie , Myocarde/métabolisme , Stress oxydatif , Facteurs de transcription/métabolisme , Séquence d'acides aminés , Animaux , Cellules COS , Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/métabolisme , Prolifération cellulaire , Chlorocebus aethiops , Protéines de liaison à l'ADN/déficit , Protéines de liaison à l'ADN/génétique , Embryon de mammifère/embryologie , Régulation de l'expression des gènes au cours du développement , Techniques de knock-out de gènes , Humains , Méthylation , Souris , Données de séquences moléculaires , Protéines du muscle/déficit , Protéines du muscle/génétique , Facteurs de transcription/déficit , Facteurs de transcription/génétique , Transcription génétique , Régulation positive
14.
Pharmacol Res ; 83: 38-51, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24373832

RÉSUMÉ

A pharmacoperone (from "pharmacological chaperone") is a small molecule that enters cells and serves as molecular scaffolding in order to cause otherwise-misfolded mutant proteins to fold and route correctly within the cell. Pharmacoperones have broad therapeutic applicability since a large number of diseases have their genesis in the misfolding of proteins and resultant misrouting within the cell. Misrouting may result in loss-of-function and, potentially, the accumulation of defective mutants in cellular compartments. Most known pharmacoperones were initially derived from receptor antagonist screens and, for this reason, present a complex pharmacology, although these are highly target specific. In this summary, we describe efforts to produce high throughput screens that identify these molecules from chemical libraries as well as a mouse model which provides proof-of-principle for in vivo protein rescue using existing pharmacoperones.


Sujet(s)
Évaluation préclinique de médicament , Tests de criblage à haut débit , Protéines/métabolisme , Bibliothèques de petites molécules/pharmacologie , Animaux , Évaluation préclinique de médicament/méthodes , Tests de criblage à haut débit/méthodes , Humains , Transport des protéines/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/composition chimique
15.
Proc Natl Acad Sci U S A ; 110(52): 21030-5, 2013 Dec 24.
Article de Anglais | MEDLINE | ID: mdl-24324164

RÉSUMÉ

Mutations in receptors, ion channels, and enzymes are frequently recognized by the cellular quality control system as misfolded and retained in the endoplasmic reticulum (ER) or otherwise misrouted. Retention results in loss of function at the normal site of biological activity and disease. Pharmacoperones are target-specific small molecules that diffuse into cells and serve as folding templates that enable mutant proteins to pass the criteria of the quality control system and route to their physiologic site of action. Pharmacoperones of the gonadotropin releasing hormone receptor (GnRHR) have efficacy in cell culture systems, and their cellular and biochemical mechanisms of action are known. Here, we show the efficacy of a pharmacoperone drug in a small animal model, a knock-in mouse, expressing a mutant GnRHR. This recessive mutation (GnRHR E(90)K) causes hypogonadotropic hypogonadism (failed puberty associated with low or apulsatile luteinizing hormone) in both humans and in the mouse model described. We find that pulsatile pharmacoperone therapy restores E(90)K from ER retention to the plasma membrane, concurrently with responsiveness to the endogenous natural ligand, gonadotropin releasing hormone, and an agonist that is specific for the mutant. Spermatogenesis, proteins associated with steroid transport and steroidogenesis, and androgen levels were restored in mutant male mice following pharmacoperone therapy. These results show the efficacy of pharmacoperone therapy in vivo by using physiological, molecular, genetic, endocrine and biochemical markers and optimization of pulsatile administration. We expect that this newly appreciated approach of protein rescue will benefit other disorders sharing pathologies based on misrouting of misfolded protein mutants.


Sujet(s)
Hypogonadisme/traitement médicamenteux , Chaperons moléculaires/pharmacologie , Pliage des protéines/effets des médicaments et des substances chimiques , Troubles de l'homéostasie des protéines/génétique , Récepteurs à la gonadolibérine/génétique , Testicule/physiologie , Animaux , Marqueurs biologiques/métabolisme , Réticulum endoplasmique/métabolisme , Techniques de knock-in de gènes , Hypogonadisme/génétique , Mâle , Souris , Chaperons moléculaires/usage thérapeutique , Mutation/génétique , Testicule/effets des médicaments et des substances chimiques
16.
Int J Biol Sci ; 8(8): 1178-87, 2012.
Article de Anglais | MEDLINE | ID: mdl-23091415

RÉSUMÉ

The distal-less homeobox gene 4 (DLX4) is a member of the DLX family of homeobox genes. Although absent from most normal adult tissues, DLX4 is widely expressed in leukemia, lung, breast, ovarian and prostate cancers. However the molecular targets, mechanisms and pathways that mediate the role of DLX4 in tumor metastasis are poorly understood. In this study, we found that DLX4 induces cancer cells to undergo epithelial to mesenchymal transition (EMT) through TWIST. Overexpression of DLX4 increased expression of TWIST expression in cancer cell lines, resulting in increased migratory and invasive capacity. Likewise, knocking down expression of DLX4 decreased TWIST expression and the migration ability of cancer cell lines. DLX4 bound to regulatory regions of the TWIST gene. Both western blotting and immunohistochemistry staining showed that the expression of DLX4 and TWIST are correlated in most of breast tumors. Taken together, these data from both cell models and tumor tissues demonstrate that DLX4 not only upregulates TWIST expression but also induces EMT and tumor metastasis. Altogether, we propose a new pathway in which DLX4 drives expression of TWIST to promote EMT, cancer migration, invasion and metastasis.


Sujet(s)
Tumeurs du sein/métabolisme , Mouvement cellulaire/physiologie , Transition épithélio-mésenchymateuse/physiologie , Régulation de l'expression des gènes tumoraux/physiologie , Protéines à homéodomaine/métabolisme , Invasion tumorale/physiopathologie , Métastase tumorale/physiopathologie , Facteurs de transcription/métabolisme , Protéine-1 apparentée à Twist/métabolisme , Technique de Western , Prolifération cellulaire , Immunoprécipitation de la chromatine , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Cellules HEK293 , Humains , Immunohistochimie , Luciferases , Interférence par ARN
17.
Mol Endocrinol ; 26(11): 1847-56, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22918878

RÉSUMÉ

GnRH, produced in the hypothalamus, acts on pituitary gonadotropes to stimulate release of the gonadotropins LH and FSH. Reduced responsiveness of gonadotropes to GnRH is a primary cause of hypogonadotropic hypogonadism (HH), a disease characterized by gonadal dysfunction and low blood levels of gonadotropins. Loss-of-function mutations in the gene encoding the receptor for GnRH (GNRHR) are a common cause of HH. Sequencing of the GNRHR gene in patients with HH revealed mainly point mutations producing single amino acid substitutions that cause misfolding and misrouting of this G protein-coupled receptor. To generate a mouse model that mimics the human disease, we introduced a single amino acid substitution (E90K) into the mouse Gnrhr gene, which is identical to a known human recessive mutation. In humans, E90K causes severe HH by preventing formation of the E90-K121 salt bridge, which is essential for correct folding. In cell cultures, E90K causes misfolding that leads to almost complete retention by the protein quality control system and subsequent degradation. Here we report that the primary phenotype of mice homozygous for E90K is female infertility due to ovulation failure. Mutant males are fertile despite reduced gonadotropin levels and smaller testes. These results suggest decreased GnRH receptor signaling in the mutant animal, compared with wild type. Our findings suggest that a threshold level of GnRH receptor activity is required for ovulation.


Sujet(s)
Anovulation/génétique , Hypogonadisme/génétique , Mutation/génétique , Troubles de l'homéostasie des protéines/génétique , Récepteurs à la gonadolibérine/génétique , Testicule/anatomopathologie , Substitution d'acide aminé/génétique , Animaux , Anovulation/sang , Anovulation/anatomopathologie , Anovulation/physiopathologie , Séquence nucléotidique , Cycle oestral , Femelle , Régulation de l'expression des gènes , Gonadotrophines/sang , Homozygote , Humains , Hypogonadisme/sang , Hypogonadisme/anatomopathologie , Hypogonadisme/physiopathologie , Lutéinisation , Mâle , Souris , Souris de lignée C57BL , Souches mutantes de souris , Données de séquences moléculaires , Taille d'organe , Récepteurs à la gonadolibérine/agonistes , Récepteurs à la gonadolibérine/métabolisme , Testicule/métabolisme
18.
Biol Reprod ; 85(5): 954-64, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21734259

RÉSUMÉ

Uterine gland formation occurs postnatally in an ovary- and steroid-independent manner in many species, including humans. Uterine glands secrete substances that are essential for embryo survival. Disruption of gland development during the postnatal period prevents gland formation, resulting in infertility. Interestingly, stabilization of beta-catenin (CTNNB1) in the uterine stroma causes a delay in gland formation rather than a complete absence of uterine glands. Thus, to determine if a critical postnatal window for gland development exists in mice, we tested the effects of extending the endocrine environment of pregnancy on uterine gland formation by treating neonatal mice with estradiol, progesterone, or oil for 5 days. One uterine horn was removed before puberty, and the other was collected at maturity. Some mice were also ovariectomized before puberty. The hormone-treated mice exhibited a delay in uterine gland formation. Hormone-treatment increased the abundance of uterine CTNNB1 and estrogen receptor alpha (ESR1) before puberty, indicating possible mechanisms for delayed gland formation. Despite having fewer glands, progesterone-treated mice were fertile, suggesting that a threshold number of glands is required for pregnancy. Mice that were ovariectomized before puberty did not undergo further uterine growth or gland development. Finally, to establish the role of the ovary in postpartum uterine gland regeneration, mice were either ovariectomized or given a sham surgery after parturition, and uteri were evaluated 1 wk later. We found that the ovary is not required for uterine growth or gland development following parturition. Thus, uterine gland development occurs continuously in mice and requires the ovary after puberty, but not after parturition.


Sujet(s)
Système génital de la femme/croissance et développement , Ovaire/physiologie , Parturition/physiologie , Maturation sexuelle/physiologie , Utérus/croissance et développement , Animaux , Animaux nouveau-nés , Oestradiol/pharmacologie , Récepteur alpha des oestrogènes/métabolisme , Femelle , Système génital de la femme/effets des médicaments et des substances chimiques , Système génital de la femme/métabolisme , Souris , Ovariectomie , Progestérone/pharmacologie , Utérus/effets des médicaments et des substances chimiques , Utérus/métabolisme , bêta-Caténine/métabolisme
19.
Biol Reprod ; 84(3): 613-8, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-21148107

RÉSUMÉ

In sexual species, fertilization of oocytes produces individuals with alleles derived from both parents. Here we use pluripotent stem cells derived from somatic cells to combine the haploid genomes from two males to produce viable sons and daughters. Male (XY) mouse induced pluripotent stem cells (Father #1) were used to isolate subclones that had spontaneously lost the Y chromosome to become genetically female (XO). These male-derived XO stem cells were used to generate female chimeras that were bred with genetically distinct males (Father #2), yielding progeny possessing genetic information that was equally derived from both fathers. Thus, functional oocytes can be generated from male somatic cells after reprogramming and spontaneous sex reversal. These findings have novel implications for mammalian reproduction and assisted reproductive technology.


Sujet(s)
Chimère/embryologie , Clonage d'organisme/méthodes , Pères , Animaux , Survie cellulaire , Cellules cultivées , Chimère/génétique , Embryon de mammifère , Femelle , Cellules souches pluripotentes induites/physiologie , Cellules souches pluripotentes induites/transplantation , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Modèles biologiques , Techniques de transfert nucléaire , Caractères sexuels
20.
Biol Reprod ; 84(2): 342-50, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-20944081

RÉSUMÉ

Sertoli cells are the primary structural component of the fetal testis cords and postnatal seminiferous tubules. Live imaging technologies facilitate the visualization of cell morphologies and behaviors through developmental processes. A transgenic mouse line was generated using a fragment of the rat Gata4 gene to direct the expression of a dual-color fluorescent protein reporter in fetal and adult Sertoli cells. The reporter encoded a red fluorescent protein, monomeric Cherry (mCherry), fused to histone 2B and enhanced green fluorescent protein (EGFP) fused to a glycosylphosphatidylinositol sequence, with a self-cleaving 2A polypeptide separating the two fusion proteins. After translation, the red and green fluorescent proteins translocated to the nucleus and plasma membrane, respectively, of Sertoli cells. Transgene expression in testes was first detected by fluorescent microscopy around Embryonic Day 12.0. Sertoli cell division and migration were visualized during testis cord formation in organ culture. Initially, the Sertoli cells had mesenchyme-like morphologies and behaviors, but later, the cells migrated to the periphery of the testis cords to become epithelialized. In postnatal seminiferous tubules, Sertoli nuclei were evenly spaced when viewed from the external surface of tubules, and Sertoli cytoplasm and membranes were associated with germ cells basally in a rosette pattern. This mouse line was bred to previously described transgenic mouse lines expressing EGFP in Sertoli cytoplasm or a nuclear cyan fluorescent protein (Cerulean) and mCherry in plasma membranes of germ cells. This revealed the physical relationship between Sertoli and germ cells in developing testis cords and provided a novel perspective on Sertoli cell development.


Sujet(s)
Animaux nouveau-nés/physiologie , Développement foetal , Foetus/cytologie , Canalicules séminifères/cytologie , Cellules de Sertoli/physiologie , Testicule/embryologie , Animaux , Membrane cellulaire/physiologie , Membrane cellulaire/ultrastructure , Cytoplasme/ultrastructure , Embryon de mammifère/métabolisme , Technique d'immunofluorescence , Colorants fluorescents , Facteur de transcription GATA-4/génétique , Expression des gènes , Gènes rapporteurs , Âge gestationnel , Protéines à fluorescence verte/génétique , Protéines luminescentes/génétique , Mâle , Souris , Souris transgéniques , Microscopie de fluorescence , Techniques de culture d'organes , Rats , Facteur de transcription SOX-9/métabolisme , Spermatozoïdes/cytologie , Testicule/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE