Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41
Filtrer
1.
Nat Commun ; 15(1): 5629, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965223

RÉSUMÉ

Mutations that decrease or increase the activity of the tyrosine phosphatase, SHP2 (encoded by PTPN11), promotes developmental disorders and several malignancies by varying phosphatase activity. We uncovered that SHP2 is a distinct class of an epigenetic enzyme; upon phosphorylation by the kinase ACK1/TNK2, pSHP2 was escorted by androgen receptor (AR) to chromatin, erasing hitherto unidentified pY54-H3 (phosphorylation of histones H3 at Tyr54) epigenetic marks to trigger a transcriptional program of AR. Noonan Syndrome with Multiple Lentigines (NSML) patients, SHP2 knock-in mice, and ACK1 knockout mice presented dramatic increase in pY54-H3, leading to loss of AR transcriptome. In contrast, prostate tumors with high pSHP2 and pACK1 activity exhibited progressive downregulation of pY54-H3 levels and higher AR expression that correlated with disease severity. Overall, pSHP2/pY54-H3 signaling acts as a sentinel of AR homeostasis, explaining not only growth retardation, genital abnormalities and infertility among NSML patients, but also significant AR upregulation in prostate cancer patients.


Sujet(s)
Épigenèse génétique , Histone , Homéostasie , Souris knockout , Tumeurs de la prostate , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Récepteurs aux androgènes , Animaux , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Histone/métabolisme , Mâle , Humains , Souris , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Phosphorylation , Syndrome de Noonan/génétique , Syndrome de Noonan/métabolisme , Transduction du signal , Chromatine/métabolisme
2.
Article de Anglais | MEDLINE | ID: mdl-38908907

RÉSUMÉ

Demodex mites are a common ectoparasite in nonlaboratory Mus musculus (mouse) populations. While infrequently reported in laboratory research mice, the prevalence is thought to be as high as 35% of all colonies. Here, we discuss an outbreak of Demodex within an SPF high-barrier vivarium housing laboratory mice first identified through commercial sentinel-free PCR testing. Consequently, in-house PCR-mediated identification of individually infected cages was conducted, and a successful method for eradication of secondary reemergent infection was generated via recurrent testing and empirical 12-wk treatment with 3 mg/kg moxidectin and 13 mg/kg imidacloprid. While we were unable to determine the source of our primary outbreak, the secondary outbreak was traced to nongenetically modified C57B6/J immunocompetent mice, which were capable of harboring subclinical infection below our PCR threshold. Our eventual successful eradication of Demodex confirmed, first, that in-house PCR detection is a cost-effective means of monitoring an outbreak; second, that treatment with 3 mg/kg moxidectin and 13 mg/kg imidacloprid does kill Demodex mites in laboratory mice; and third, that treatment of only PCR-positive mice is an insufficient way to control an outbreak. Taken together, our methodological approach for infestations such as Demodex suggests it is possible to eradicate them but that it requires a thorough, systematic, and aggressive treatment regimen. Moreover, we recommend that all cages derived from infected animals be treated as positive, regardless of PCR positivity, to prevent recurrent and/or persistent infections within an animal colony.

3.
Adv Sci (Weinh) ; : e2308975, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38757640

RÉSUMÉ

Over the past decades, mesenchymal stromal cells (MSCs) have been extensively investigated as a potential therapeutic cell source for the treatment of various disorders. Differentiation of MSCs from human induced pluripotent stem cells (iMSCs) has provided a scalable approach for the biomanufacturing of MSCs and related biological products. Although iMSCs shared typical MSC markers and functions as primary MSCs (pMSCs), there is a lack of lineage specificity in many iMSC differentiation protocols. Here, a stepwise hiPSC-to-iMSC differentiation method is employed via intermediate cell stages of neural crest and cytotrophoblast to generate lineage-specific MSCs with varying differentiation efficiencies and gene expression. Through a comprehensive comparison between early developmental cell types (hiPSCs, neural crest, and cytotrophoblast), two lineage-specific iMSCs, and six source-specific pMSCs, are able to not only distinguish the transcriptomic differences between MSCs and early developmental cells, but also determine the transcriptomic similarities of iMSC subtypes to postnatal or perinatal pMSCs. Additionally, it is demonstrated that different iMSC subtypes and priming conditions affected EV production, exosomal protein expression, and cytokine cargo.

4.
Article de Anglais | MEDLINE | ID: mdl-38489782

RÉSUMÉ

Cardiovascular disease (CVD) remains the number one cause of death worldwide. Women are at increased risk of death from CVD, but the mechanisms for how and why this occurs remain elusive. One subset of women who are exceptionally vulnerable to CVD are those with rheumatic diseases (RDs). Indeed, women account for 80% of all RDs, disorders that encompass a broad range of autoimmune and autoinflammatory diseases that lead to chronic inflammation and pathology. The clear association of increased CVD risk in women with RD is thought to be mediated by a number of factors, including RD pathology itself, pharmacological induction of CVD, and/or as yet unidentified mechanisms. As such, elucidation of the causes and treatments of these pathologies has given rise to a new subspecialty of cardiology: cardio-rheumatology. Here, we review and discuss the CVD risks in patients with RDs, the associated sex disparities in RD and CVD care, as well as the current therapeutic and interventional options available to specifically help women with RDs. We hope this discussion will provide guidance and support to patients, as well as to cardio-rheumatologists, as these groups are the most uniquely positioned to radically improve CVD care in these individuals. Moreover, we are hopeful this discussion may lead to better, more efficacious approaches to treating these disorders in women in the near future.

5.
Am J Med Genet A ; 194(4): e63477, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37969032

RÉSUMÉ

Germline pathogenic variants in the RAS/mitogen-activated protein kinase (MAPK) signaling pathway are the molecular cause of RASopathies, a group of clinically overlapping genetic syndromes. RASopathies constitute a wide clinical spectrum characterized by distinct facial features, short stature, predisposition to cancer, and variable anomalies in nearly all the major body systems. With increasing global recognition of these conditions, the 8th International RASopathies Symposium spotlighted global perspectives on clinical care and research, including strategies for building international collaborations and developing diverse patient cohorts in anticipation of interventional trials. This biannual meeting, organized by RASopathies Network, was held in a hybrid virtual/in-person format. The agenda featured emerging discoveries and case findings as well as progress in preclinical and therapeutic pipelines. Stakeholders including basic scientists, clinician-scientists, practitioners, industry representatives, patients, and family advocates gathered to discuss cutting edge science, recognize current gaps in knowledge, and hear from people with RASopathies about the experience of daily living. Presentations by RASopathy self-advocates and early-stage investigators were featured throughout the program to encourage a sustainable, diverse, long-term research and advocacy partnership focused on improving health and bringing treatments to people with RASopathies.


Sujet(s)
Syndrome de Costello , Dysplasie ectodermique , Cardiopathies congénitales , Tumeurs , Syndrome de Noonan , Humains , Protéines G ras/génétique , Système de signalisation des MAP kinases/génétique , Syndrome de Costello/génétique , Tumeurs/génétique , Dysplasie ectodermique/génétique , Syndrome de Noonan/génétique , Cardiopathies congénitales/génétique
6.
Adv Mater ; 36(8): e2304615, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37934471

RÉSUMÉ

The spleen is an important mediator of both adaptive and innate immunity. As such, attempts to modulate the immune response provided by the spleen may be conducive to improved outcomes for numerous diseases throughout the body. Here, biomimicry is used to rationally design nanomaterials capable of splenic retention and immunomodulation for the treatment of disease in a distant organ, the postinfarct heart. Engineered senescent erythrocyte-derived nanotheranostic (eSENTs) are generated, demonstrating significant uptake by the immune cells of the spleen including T and B cells, as well as monocytes and macrophages. When loaded with suberoylanilide hydroxamic acid (SAHA), the nanoagents exhibit a potent therapeutic effect, reducing infarct size by 14% at 72 h postmyocardial infarction when given as a single intravenous dose 2 h after injury. These results are supportive of the hypothesis that RBC-derived biomimicry may provide new approaches for the targeted modulation of the pathological processes involved in myocardial infarction, thus further experiments to decisively confirm the mechanisms of action are currently underway. This novel concept may have far-reaching applicability for the treatment of a number of both acute and chronic conditions where the immune responses are either stimulated or suppressed by the splenic (auto)immune milieu.


Sujet(s)
Biomimétique , Infarctus du myocarde , Humains , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Coeur , Immunité innée , Immunomodulation
7.
J Mol Cell Cardiol ; 175: 62-66, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36584478

RÉSUMÉ

Myh6-Cre transgenic mouse line was known to express Cre recombinase only in the heart. Nevertheless, during breeding Myh6-Cre to Rosa26fstdTom reporter (tdTom) mouse line, we observed that a significant part of their F2 tdTom/+ offspring had tdTom reporter gene universally activated. Our results show that Myh6-Cre transgenic mice have Cre recombinase activity in a subpopulation of the male germline cells, and that Myh6 gene transcripts are enriched in the interstitial Leydig cells and the undifferentiated spermatogonia stem cells. In summary, the current study confirms that the previously known "heart-specific" Myh6 promoter drives Cre expression in the testis.


Sujet(s)
Cellules germinales , Integrases , Mâle , Souris , Animaux , Régions promotrices (génétique)/génétique , Souris transgéniques , Integrases/génétique , Integrases/métabolisme , Cellules germinales/métabolisme
8.
J Clin Invest ; 132(8): 1-5, 2022 04 15.
Article de Anglais | MEDLINE | ID: mdl-35426371

RÉSUMÉ

RASopathies are a family of rare autosomal dominant disorders that affect the canonical Ras/MAPK signaling pathway and manifest as neurodevelopmental systemic syndromes, including Costello syndrome (CS). In this issue of the JCI, Dard et al. describe the molecular determinants of CS using a myriad of genetically modified models, including mice expressing HRAS p.G12S, patient-derived skin fibroblasts, hiPSC-derived human cardiomyocytes, an HRAS p.G12V zebrafish model, and human lentivirally induced fibroblasts overexpressing HRAS p.G12S or HRAS p.G12A. Mitochondrial proteostasis and oxidative phosphorylation were altered in CS, and inhibition of the AMPK signaling pathway mediated bioenergetic changes. Importantly, the pharmacological induction of this pathway restored cardiac function and reduced the developmental defects associated with CS. These findings identify a role for altered bioenergetics and provide insights into more effective treatment strategies for patients with RASopathies.


Sujet(s)
Syndrome de Costello , Danio zébré , Animaux , Syndrome de Costello/métabolisme , Métabolisme énergétique , Humains , Souris , Mitochondries/génétique , Mitochondries/métabolisme , Protéines proto-oncogènes p21(ras)/métabolisme , Transduction du signal , Danio zébré/métabolisme
9.
Adv Sci (Weinh) ; 9(17): e2200829, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35373532

RÉSUMÉ

The cellular response to stress is an important determinant of disease pathogenesis. Uncovering the molecular fingerprints of distinct stress responses may identify novel biomarkers and key signaling pathways for different diseases. Emerging evidence shows that transfer RNA-derived small RNAs (tDRs) play pivotal roles in stress responses. However, RNA modifications present on tDRs are barriers to accurately quantifying tDRs using traditional small RNA sequencing. Here, AlkB-facilitated methylation sequencing is used to generate a comprehensive landscape of cellular and extracellular tDR abundances in various cell types during different stress responses. Extracellular tDRs are found to have distinct fragmentation signatures from intracellular tDRs and these tDR signatures are better indicators of different stress responses than miRNAs. These distinct extracellular tDR fragmentation patterns and signatures are also observed in plasma from patients on cardiopulmonary bypass. It is additionally demonstrated that angiogenin and RNASE1 are themselves regulated by stressors and contribute to the stress-modulated abundance of sub-populations of cellular and extracellular tDRs. Finally, a sub-population of extracellular tDRs is identified for which AGO2 appears to be required for their expression. Together, these findings provide a detailed profile of stress-responsive tDRs and provide insight about tDR biogenesis and stability in response to cellular stressors.


Sujet(s)
microARN , ARN de transfert , Séquence nucléotidique , Humains , microARN/génétique , ARN de transfert/génétique , ARN de transfert/métabolisme , Analyse de séquence d'ARN
11.
Am J Med Genet A ; 188(6): 1915-1927, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35266292

RÉSUMÉ

RASopathies are a group of genetic disorders that are caused by genes that affect the canonical Ras/mitogen-activated protein kinase (MAPK) signaling pathway. Despite tremendous progress in understanding the molecular consequences of these genetic anomalies, little movement has been made in translating these findings to the clinic. This year, the seventh International RASopathies Symposium focused on expanding the research knowledge that we have gained over the years to enhance new discoveries in the field, ones that we hope can lead to effective therapeutic treatments. Indeed, for the first time, research efforts are finally being translated to the clinic, with compassionate use of Ras/MAPK pathway inhibitors for the treatment of RASopathies. This biannual meeting, organized by the RASopathies Network, brought together basic scientists, clinicians, clinician scientists, patients, advocates, and their families, as well as representatives from pharmaceutical companies and the National Institutes of Health. A history of RASopathy gene discovery, identification of new disease genes, and the latest research, both at the bench and in the clinic, were discussed.


Sujet(s)
Syndrome de Costello , Syndrome de Noonan , Syndrome de Costello/génétique , Humains , Mitogen-Activated Protein Kinases/métabolisme , Syndrome de Noonan/génétique , Transduction du signal , Protéines G ras/génétique , Protéines G ras/métabolisme
12.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-34206257

RÉSUMÉ

Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) that modulate innate immune responses and play essential roles in the pathogenesis of heart diseases. Although important, the molecular mechanisms controlling cardiac TLR genes expression have not been clearly addressed. This study examined the expression pattern of Tlr1, Tlr2, Tlr3, Tlr4, Tlr5, Tlr6, Tlr7, Tlr8, and Tlr9 in normal and disease-stressed mouse hearts. Our results demonstrated that the expression levels of cardiac Tlr3, Tlr7, Tlr8, and Tlr9 increased with age between neonatal and adult developmental stages, whereas the expression of Tlr5 decreased with age. Furthermore, pathological stress increased the expression levels of Tlr2, Tlr4, Tlr5, Tlr7, Tlr8, and Tlr9. Hippo-YAP signaling is essential for heart development and homeostasis maintenance, and YAP/TEAD1 complex is the terminal effector of this pathway. Here we found that TEAD1 directly bound genomic regions adjacent to Tlr1, Tlr2, Tlr3, Tlr4, Tlr5, Tlr6, Tlr7, and Tlr9. In vitro, luciferase reporter data suggest that YAP/TEAD1 repression of Tlr4 depends on a conserved TEAD1 binding motif near Tlr4 transcription start site. In vivo, cardiomyocyte-specific YAP depletion increased the expression of most examined TLR genes, activated the synthesis of pro-inflammatory cytokines, and predisposed the heart to lipopolysaccharide stress. In conclusion, our data indicate that the expression of cardiac TLR genes is associated with age and activated by pathological stress and suggest that YAP/TEAD1 complex is a default repressor of cardiac TLR genes.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines de liaison à l'ADN/métabolisme , Immunité innée , Myocytes cardiaques/métabolisme , Récepteurs de type Toll/génétique , Facteurs de transcription/métabolisme , Facteurs âges , Animaux , Cytokines/métabolisme , Régulation de l'expression des gènes , Lipopolysaccharides , Souris , Souris de lignée C57BL , Transduction du signal , Facteurs de transcription à domaine TEA , Protéines de signalisation YAP
13.
Stem Cell Reports ; 16(5): 1228-1244, 2021 05 11.
Article de Anglais | MEDLINE | ID: mdl-33891865

RÉSUMÉ

Emerging technologies in stem cell engineering have produced sophisticated organoid platforms by controlling stem cell fate via biomaterial instructive cues. By micropatterning and differentiating human induced pluripotent stem cells (hiPSCs), we have engineered spatially organized cardiac organoids with contracting cardiomyocytes in the center surrounded by stromal cells distributed along the pattern perimeter. We investigated how geometric confinement directed the structural morphology and contractile functions of the cardiac organoids and tailored the pattern geometry to optimize organoid production. Using modern data-mining techniques, we found that pattern sizes significantly affected contraction functions, particularly in the parameters related to contraction duration and diastolic functions. We applied cardiac organoids generated from 600 µm diameter circles as a developmental toxicity screening assay and quantified the embryotoxic potential of nine pharmaceutical compounds. These cardiac organoids have potential use as an in vitro platform for studying organoid structure-function relationships, developmental processes, and drug-induced cardiac developmental toxicity.


Sujet(s)
Développement embryonnaire , Coeur/embryologie , Organoïdes/embryologie , Ingénierie tissulaire , Tests de toxicité , Signalisation calcique , Différenciation cellulaire , Coeur/physiologie , Humains , Cellules souches pluripotentes induites/cytologie , Organoïdes/physiologie
14.
Am J Med Genet A ; 182(3): 597-606, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31825160

RÉSUMÉ

The RASopathies are a group of genetic disorders that result from germline pathogenic variants affecting RAS-mitogen activated protein kinase (MAPK) pathway genes. RASopathies share RAS/MAPK pathway dysregulation and share phenotypic manifestations affecting numerous organ systems, causing lifelong and at times life-limiting medical complications. RASopathies may benefit from precision medicine approaches. For this reason, the Sixth International RASopathies Symposium focused on exploring precision medicine. This meeting brought together basic science researchers, clinicians, clinician scientists, patient advocates, and representatives from pharmaceutical companies and the National Institutes of Health. Novel RASopathy genes, variants, and animal models were discussed in the context of medication trials and drug development. Attempts to define and measure meaningful endpoints for treatment trials were discussed, as was drug availability to patients after trial completion.


Sujet(s)
Maladies génétiques congénitales/génétique , Mitogen-Activated Protein Kinase Kinases/génétique , Protéines G ras/génétique , Maladies génétiques congénitales/anatomopathologie , Mutation germinale/génétique , Humains , Transduction du signal/génétique
15.
Circulation ; 140(3): 207-224, 2019 07 16.
Article de Anglais | MEDLINE | ID: mdl-31163979

RÉSUMÉ

BACKGROUND: More than 90% of individuals with Noonan syndrome (NS) with mutations clustered in the CR2 domain of RAF1 present with severe and often lethal hypertrophic cardiomyopathy (HCM). The signaling pathways by which NS RAF1 mutations promote HCM remain elusive, and so far, there is no known treatment for NS-associated HCM. METHODS: We used patient-derived RAF1S257L/+ and CRISPR-Cas9-generated isogenic control inducible pluripotent stem cell (iPSC)-derived cardiomyocytes to model NS RAF1-associated HCM and to further delineate the molecular mechanisms underlying the disease. RESULTS: We show that mutant iPSC-derived cardiomyocytes phenocopy the pathology seen in hearts of patients with NS by exhibiting hypertrophy and structural defects. Through pharmacological and genetic targeting, we identify 2 perturbed concomitant pathways that, together, mediate HCM in RAF1 mutant iPSC-derived cardiomyocytes. Hyperactivation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), but not extracellular regulated kinase 1/2, causes myofibrillar disarray, whereas the enlarged cardiomyocyte phenotype is a direct consequence of increased extracellular regulated kinase 5 (ERK5) signaling, a pathway not previously known to be involved in NS. RNA-sequencing reveals genes with abnormal expression in RAF1 mutant iPSC-derived cardiomyocytes and identifies subsets of genes dysregulated by aberrant MEK1/2 or ERK5 pathways that could contribute to the NS-associated HCM. CONCLUSIONS: Taken together, the results of our study identify the molecular mechanisms by which NS RAF1 mutations cause HCM and reveal downstream effectors that could serve as therapeutic targets for treatment of NS and perhaps other, more common, congenital HCM disorders.


Sujet(s)
Cardiomyopathie hypertrophique/génétique , Cellules souches pluripotentes induites/physiologie , MAP Kinase Kinase 1/génétique , MAP Kinase Kinase 2/génétique , Mitogen-Activated Protein Kinase 7/génétique , Syndrome de Noonan/génétique , Protéines proto-oncogènes c-raf/génétique , Adolescent , Systèmes CRISPR-Cas/physiologie , Cardiomyopathie hypertrophique/métabolisme , Cellules cultivées , Enfant , Femelle , Cellules HEK293 , Humains , MAP Kinase Kinase 1/métabolisme , MAP Kinase Kinase 2/métabolisme , Mâle , Mitogen-Activated Protein Kinase 7/métabolisme , Myocytes cardiaques/physiologie , Syndrome de Noonan/métabolisme , Protéines proto-oncogènes c-raf/métabolisme
16.
Stem Cell Res ; 34: 101374, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30640061

RÉSUMÉ

Noonan syndrome with multiple lentigines (NSML), formerly known as LEOPARD Syndrome, is a rare autosomal dominant disorder. Approximately 90% of NSML cases are caused by missense mutations in the PTPN11 gene which encodes the protein tyrosine phosphatase SHP2. A human induced pluripotent stem cell (iPSC) line was generated using peripheral blood mononuclear cells (PBMCs) from a patient with NSML that carries a gene mutation of p.Q510P on the PTPN11 gene using non-integrating Sendai virus technique. This iPSC line offers a useful resource to study the disease pathophysiology and a cell-based model for drug development to treat NSML.


Sujet(s)
Techniques de culture cellulaire/méthodes , Cellules souches pluripotentes induites/anatomopathologie , Syndrome LEOPARD/génétique , Syndrome LEOPARD/anatomopathologie , Mutation/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Adolescent , Séquence nucléotidique , Lignée cellulaire , Femelle , Humains
17.
Curr Opin Physiol ; 1: 123-139, 2018 Feb.
Article de Anglais | MEDLINE | ID: mdl-29532042

RÉSUMÉ

The heart is one of the first organs to form and function during embryonic development. It is comprised of multiple cell lineages, each integral for proper cardiac development, and include cardiomyocytes, endothelial cells, epicardial cells and neural crest cells. The molecular mechanisms regulating cardiac development and morphogenesis are dependent on signaling crosstalk between multiple lineages through paracrine interactions, cell-ECM interactions, and cell-cell interactions, which together, help facilitate survival, growth, proliferation, differentiation and migration of cardiac tissue. Aberrant regulation of any of these processes can induce developmental disorders and pathological phenotypes. Here, we will discuss each of these processes, the genetic factors that contribute to each step of cardiac development, as well as the current and future therapeutic targets and mechanisms of heart development and disease. Understanding the complex interactions that regulate cardiac development, proliferation and differentiation is not only vital to understanding the causes of congenital heart defects, but to also finding new therapeutics that can treat both pediatric and adult cardiac disease in the near future.

18.
Sci Signal ; 11(522)2018 03 20.
Article de Anglais | MEDLINE | ID: mdl-29559584

RÉSUMÉ

Catalytically activating mutations in Ptpn11, which encodes the protein tyrosine phosphatase SHP2, cause 50% of Noonan syndrome (NS) cases, whereas inactivating mutations in Ptpn11 are responsible for nearly all cases of the similar, but distinct, developmental disorder Noonan syndrome with multiple lentigines (NSML; formerly called LEOPARD syndrome). However, both types of disease mutations are gain-of-function mutations because they cause SHP2 to constitutively adopt an open conformation. We found that the catalytic activity of SHP2 was required for the pathogenic effects of gain-of-function, disease-associated mutations on the development of hydrocephalus in the mouse. Targeted pan-neuronal knockin of a Ptpn11 allele encoding the active SHP2 E76K mutant resulted in hydrocephalus due to aberrant development of ependymal cells and their cilia. These pathogenic effects of the E76K mutation were suppressed by the additional mutation C459S, which abolished the catalytic activity of SHP2. Moreover, ependymal cells in NSML mice bearing the inactive SHP2 mutant Y279C were also unaffected. Mechanistically, the SHP2 E76K mutant induced developmental defects in ependymal cells by enhancing dephosphorylation and inhibition of the transcription activator STAT3. Whereas STAT3 activity was reduced in Ptpn11E76K/+ cells, the activities of the kinases ERK and AKT were enhanced, and neural cell-specific Stat3 knockout mice also manifested developmental defects in ependymal cells and cilia. These genetic and biochemical data demonstrate a catalytic-dependent role of SHP2 gain-of-function disease mutants in the pathogenesis of hydrocephalus.


Sujet(s)
Mutation gain de fonction , Hydrocéphalie/génétique , Syndrome LEOPARD/génétique , Syndrome de Noonan/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Animaux , Biocatalyse , Épendyme/cytologie , Épendyme/métabolisme , Prédisposition génétique à une maladie/génétique , Humains , Hydrocéphalie/métabolisme , Syndrome LEOPARD/métabolisme , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Cellules souches neurales/métabolisme , Syndrome de Noonan/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme
19.
PLoS One ; 12(6): e0178905, 2017.
Article de Anglais | MEDLINE | ID: mdl-28582432

RÉSUMÉ

Noonan Syndrome with Multiple Lentigines (NSML, formerly LEOPARD syndrome) is an autosomal dominant "RASopathy" disorder manifesting in congenital heart disease. Most cases of NSML are caused by catalytically inactivating mutations in the protein tyrosine phosphatase (PTP), non-receptor type 11 (PTPN11), encoding the SH2 domain-containing PTP-2 (SHP2) protein. We previously generated knock-in mice harboring the PTPN11 mutation Y279C, one of the most common NSML alleles; these now-termed SHP2Y279C/+ mice recapitulate the human disorder and develop hypertrophic cardiomyopathy (HCM) by 12 weeks of age. Functionally, heart and/or cardiomyocyte lysates from SHP2Y279C/+ mice exhibit increased basal and agonist-induced AKT and mTOR activities. Here, we sought to determine whether we could reverse the hypertrophy in SHP2Y279C/+ mice using ARQ 092, an oral and selective allosteric AKT inhibitor currently in clinical trials for patients with PI3K/AKT-driven tumors or Proteus syndrome. We obtained echocardiographs of SHP2Y279C/+ and wildtype (SHP2+/+) littermates, either in the presence or absence of ARQ 092 at 12, 14, and 16 weeks of age. While SHP2Y279C/+ mice developed significant left ventricular hypertrophy by 12 weeks, as indicated by decreased chamber dimension and increased posterior wall thickness, treatment of SHP2Y279C/+ mice with ARQ 092 normalized the hypertrophy in as early as 2 weeks following treatment, with hearts comparable in size to those in wildtype (SHP2+/+) mice. In addition, we observed an increase in fractional shortening (FS%) in SHP2Y279C/+ mice, an effect of increased compensatory hypertrophy, which was not apparent in SHP2Y279C/+ mice treated with ARQ 092, suggesting functional improvement of HCM upon treatment with the AKT inhibitor. Finally, we found that ARQ 092 specifically inhibited AKT activity, as well as its downstream effectors, PRAS and S6RP in NSML mice. Taken together, these data suggest ARQ 092 may be a promising novel therapy for treatment of hypertrophy in NSML patients.


Sujet(s)
Aminopyridines/pharmacologie , Cardiomyopathie hypertrophique/traitement médicamenteux , Cardiotoniques/pharmacologie , Imidazoles/pharmacologie , Syndrome LEOPARD/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Allèles , Animaux , Cardiomyopathie hypertrophique/imagerie diagnostique , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Modèles animaux de maladie humaine , Échocardiographie , Régulation de l'expression des gènes , Humains , Syndrome LEOPARD/imagerie diagnostique , Syndrome LEOPARD/génétique , Syndrome LEOPARD/métabolisme , Mâle , Souris , Souris transgéniques , Mutation , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Phosphoprotéines/antagonistes et inhibiteurs , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , Domaines protéiques , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/métabolisme
20.
Sci Signal ; 10(468)2017 02 28.
Article de Anglais | MEDLINE | ID: mdl-28246202

RÉSUMÉ

Physiological cardiac hypertrophy, in response to stimuli such as exercise, is considered adaptive and beneficial. In contrast, pathological cardiac hypertrophy that arises in response to pathological stimuli such as unrestrained high blood pressure and oxidative or metabolic stress is maladaptive and may precede heart failure. We found that the transcript encoding DNA damage-inducible transcript 4-like (DDiT4L) was expressed in murine models of pathological cardiac hypertrophy but not in those of physiological cardiac hypertrophy. In cardiomyocytes, DDiT4L localized to early endosomes and promoted stress-induced autophagy through a process involving mechanistic target of rapamycin complex 1 (mTORC1). Exposing cardiomyocytes to various types of pathological stress increased the abundance of DDiT4L, which inhibited mTORC1 but activated mTORC2 signaling. Mice with conditional cardiac-specific overexpression of DDiT4L had mild systolic dysfunction, increased baseline autophagy, reduced mTORC1 activity, and increased mTORC2 activity, all of which were reversed by suppression of transgene expression. Genetic suppression of autophagy also reversed cardiac dysfunction in these mice. Our data showed that DDiT4L may be an important transducer of pathological stress to autophagy through mTOR signaling in the heart and that DDiT4L could be therapeutically targeted in cardiovascular diseases in which autophagy and mTOR signaling play a major role.


Sujet(s)
Autophagie/génétique , Cardiomégalie/génétique , Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes , Facteurs de transcription/génétique , Protéines adaptatrices de la transduction du signal , Animaux , Animaux nouveau-nés , Technique de Western , Cardiomégalie/métabolisme , Cardiomégalie/anatomopathologie , Cellules cultivées , Protéines de liaison à l'ADN/métabolisme , Endosomes/génétique , Endosomes/métabolisme , Cellules HEK293 , Humains , Complexe-1 cible mécanistique de la rapamycine/génétique , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/génétique , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Souris knockout , Souris transgéniques , Microscopie confocale , Myocytes cardiaques/métabolisme , Stress oxydatif , Rats , Transduction du signal/génétique , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...