Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Biochim Biophys Acta Rev Cancer ; 1878(5): 188951, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37433417

RESUMEN

Glioma is the most prevalent type of neurological malignancies. Despite decades of efforts in neurosurgery, chemotherapy and radiation therapy, glioma remains one of the most treatment-resistant brain tumors with unfavorable outcomes. Recent progresses in genomic and epigenetic profiling have revealed new concepts of genetic events involved in the etiology of gliomas in humans, meanwhile, revolutionary technologies in gene editing and delivery allows to code these genetic "events" in animals to genetically engineer glioma models. This approach models the initiation and progression of gliomas in a natural microenvironment with an intact immune system and facilitates probing therapeutic strategies. In this review, we focus on recent advances in in vivo electroporation-based glioma modeling and outline the established genetically engineered glioma models (GEGMs).


Asunto(s)
Neoplasias Encefálicas , Glioma , Animales , Humanos , Glioma/tratamiento farmacológico , Neoplasias Encefálicas/patología , Ingeniería Genética , Electroporación , Sistema Inmunológico , Microambiente Tumoral
2.
Epilepsy Curr ; 22(4): 234-240, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187145

RESUMEN

Focal cortical dysplasia (FCD) is a malformation of cortical development that is a prevalent cause of intractable epilepsy in children. Of the three FCD subtypes, understanding the etiology and pathogenesis of FCD type II has seen the most progress owing to the recent advances in identifying gene mutations along the mTOR signaling pathway as a frequent cause of this disorder. Accordingly, numerous animal models of FCD type II based on genetic manipulation of the mTOR signaling pathway have emerged to investigate the mechanisms of epileptogenesis and novel therapeutics for epilepsy. These include transgenic and in utero electroporation-based animal models. Here, we review the histopathological and electroclinical features of existing FCD type II animal models and discuss the scientific and technical considerations, clinical applications, and limitations of current models. We also highlight other models of FCD based on early life acquired factors.

4.
Epilepsy Res ; 181: 106890, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35219048

RESUMEN

Tuberous sclerosis complex (TSC) is a monogenic disorder characterized by hyperactivation of the mTOR signaling pathway and developmental brain malformations leading to intractable epilepsy. Although treatment with the recently approved mTOR inhibitor, everolimus, results in clinically relevant seizure suppression in up to 40% of TSC patients, seizures remain uncontrolled in a large number of cases, underscoring the need to identify novel treatment targets. The MEK-ERK signaling pathway has been found to be aberrantly activated in TSC and inhibition of MEK-ERK activity independently of mTOR rescued neuronal dendrite overgrowth in mice modeling TSC neuropathology. Here, we evaluated the efficacy of MEK-ERK inhibition on seizures in two mouse models of TSC. We found that treatment with the MEK inhibitor PD0325901 (mirdametinib) significantly reduced seizure activity in both TSC mouse models. These findings support inhibiting MEK-ERK activity as a potential alternative strategy to treat seizures in TSC.


Asunto(s)
Esclerosis Tuberosa , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Convulsiones/tratamiento farmacológico , Transducción de Señal , Esclerosis Tuberosa/complicaciones , Esclerosis Tuberosa/tratamiento farmacológico
5.
JCI Insight ; 6(20)2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34676826

RESUMEN

Failed or altered gliogenesis is a major characteristic of diffuse white matter injury in survivors of premature birth. The developmentally regulated long noncoding RNA (lncRNA) H19 inhibits S-adenosylhomocysteine hydrolase (SAHH) and contributes to methylation of diverse cellular components, such as DNA, RNA, proteins, lipids, and neurotransmitters. We showed that the pregnancy-derived synthetic PreImplantation Factor (sPIF) induces expression of the nuclear receptor corepressor 2 (NCOR2) via H19/SAHH-mediated DNA demethylation. In turn, NCOR2 affects oligodendrocyte differentiation markers. Accordingly, after hypoxic-ischemic brain injury in rodents, myelin protection and oligodendrocytes' fate are in part modulated by sPIF and H19. Our results revealed an unexpected mechanism of the H19/SAHH axis underlying myelin preservation during brain recovery and its use in treating neurodegenerative diseases can be envisioned.


Asunto(s)
Co-Represor 2 de Receptor Nuclear/metabolismo , Oligodendroglía/fisiología , Péptidos/fisiología , ARN Largo no Codificante/genética , Animales , Femenino , Humanos , Ratones , Embarazo
6.
Front Neuroanat ; 15: 715363, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34295225

RESUMEN

[This corrects the article DOI: 10.3389/fnana.2021.664695.].

7.
Front Neuroanat ; 15: 664695, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897381

RESUMEN

Hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) due to mutations in genes along the PI3K-mTOR pathway and the GATOR1 complex causes a spectrum of neurodevelopmental disorders (termed mTORopathies) associated with malformation of cortical development and intractable epilepsy. Despite these gene variants' converging impact on mTORC1 activity, emerging findings suggest that these variants contribute to epilepsy through both mTORC1-dependent and -independent mechanisms. Here, we review the literature on in utero electroporation-based animal models of mTORopathies, which recapitulate the brain mosaic pattern of mTORC1 hyperactivity, and compare the effects of distinct PI3K-mTOR pathway and GATOR1 complex gene variants on cortical development and epilepsy. We report the outcomes on cortical pyramidal neuronal placement, morphology, and electrophysiological phenotypes, and discuss some of the converging and diverging mechanisms responsible for these alterations and their contribution to epileptogenesis. We also discuss potential therapeutic strategies for epilepsy, beyond mTORC1 inhibition with rapamycin or everolimus, that could offer personalized medicine based on the gene variant.

8.
Sci Transl Med ; 12(570)2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208499

RESUMEN

The causative link between focal cortical malformations (FCMs) and epilepsy is well accepted, especially among patients with focal cortical dysplasia type II (FCDII) and tuberous sclerosis complex (TSC). However, the mechanisms underlying seizures remain unclear. Using a mouse model of TSC- and FCDII-associated FCM, we showed that FCM neurons were responsible for seizure activity via their unexpected abnormal expression of the hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4 (HCN4), which is normally not present in cortical pyramidal neurons after birth. Increasing intracellular cAMP concentrations, which preferentially affects HCN4 gating relative to the other isoforms, drove repetitive firing of FCM neurons but not control pyramidal neurons. Ectopic HCN4 expression was dependent on the mechanistic target of rapamycin (mTOR), preceded the onset of seizures, and was also found in diseased neurons in tissue resected from patients with TSC and FCDII. Last, blocking HCN4 channel activity in FCM neurons prevented epilepsy in the mouse model. These findings suggest that HCN4 play a main role in seizure and identify a cAMP-dependent seizure mechanism in TSC and FCDII. Furthermore, the unique expression of HCN4 exclusively in FCM neurons suggests that gene therapy targeting HCN4 might be effective in reducing seizures in FCDII or TSC.


Asunto(s)
Epilepsia , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Malformaciones del Desarrollo Cortical de Grupo I , Esclerosis Tuberosa , Epilepsia/genética , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares , Canales de Potasio/genética , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/genética
9.
Sci Transl Med ; 12(531)2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075941

RESUMEN

Epilepsy treatments for patients with mechanistic target of rapamycin (mTOR) disorders, such as tuberous sclerosis complex (TSC) or focal cortical dysplasia type II (FCDII), are urgently needed. In these patients, the presence of focal cortical malformations is associated with the occurrence of lifelong epilepsy, leading to severe neurological comorbidities. Here, we show that the expression of the actin cross-linking protein filamin A (FLNA) is increased in resected cortical tissue that is responsible for seizures in patients with FCDII and in mice modeling TSC and FCDII with mutations in phosphoinositide 3-kinase (PI3K)-ras homolog enriched in brain (Rheb) pathway genes. Normalizing FLNA expression in these mice through genetic knockdown limited cell misplacement and neuronal dysmorphogenesis, two hallmarks of focal cortical malformations. In addition, Flna knockdown reduced seizure frequency independently of mTOR signaling. Treating mice with a small molecule targeting FLNA, PTI-125, before the onset of seizures alleviated neuronal abnormalities and reduced seizure frequency compared to vehicle-treated mice. In addition, the treatment was also effective when injected after seizure onset in juvenile and adult mice. These data suggest that targeting FLNA with either short hairpin RNAs or the small molecule PTI-125 might be effective in reducing seizures in patients with TSC and FCDII bearing mutations in PI3K-Rheb pathway genes.


Asunto(s)
Epilepsia , Malformaciones del Desarrollo Cortical de Grupo I , Animales , Filaminas , Humanos , Ratones , Fosfatidilinositol 3-Quinasas , Convulsiones/tratamiento farmacológico
10.
Epilepsia ; 60(11): 2163-2173, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31625153

RESUMEN

The mechanistic target of rapamycin (mTOR) pathway has been implicated in a growing number of malformations of cortical development (MCD) associated with intractable epilepsy. Mutations in single genes encoding mTOR pathway regulatory proteins have been linked to MCD such as focal cortical dysplasia (FCD) types IIa and IIb, hemimegalencephaly (HME), and megalencephaly. Recent studies have demonstrated that the GATOR1 protein complex, comprised of DEPDC5, NPRL3, and NPRL2, plays a pivotal role in regulating mTOR signaling in response to cellular amino acid levels and that mutations in DEPDC5, NPRL3, or NPRL2 are linked to FCD, HME, and seizures. Histopathological analysis of FCD and HME tissue specimens resected from individuals harboring DEPDC5, NPRL3, or NPRL2 gene mutations reveals hyperactivation of mTOR pathway signaling. Family pedigrees carrying mutations in either DEPDC5 or NPRL3 share clinical phenotypes of epilepsy and MCD, as well as intellectual and neuropsychiatric disabilities. Interestingly, some individuals with seizures associated with DEPDC5, NPRL3, or NPRL2 variants exhibit normal brain imaging suggesting either occult MCD or a role for these genes in non-lesional neocortical epilepsy. Mouse models resulting from knockdown or knockout of either Depdc5 or Nprl3 exhibit altered cortical lamination, neuronal dysmorphogenesis, and enhanced neuronal excitability as reported in models resulting from direct mTOR activation through expression of its canonical activator RHEB. The role of the GATOR1 proteins in regulating mTOR signaling suggest plausible options for mTOR inhibition in the treatment of epilepsy associated with mutations in DEPDC5, NPRL3, or NPRL2.


Asunto(s)
Epilepsia/genética , Malformaciones del Desarrollo Cortical/genética , Mutación/genética , Serina-Treonina Quinasas TOR/genética , Animales , Epilepsia/diagnóstico por imagen , Proteínas Activadoras de GTPasa/genética , Humanos , Malformaciones del Desarrollo Cortical/diagnóstico por imagen , Proteínas Supresoras de Tumor/genética
11.
Epilepsia ; 60(6): 1255-1265, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31125447

RESUMEN

OBJECTIVES: Patients with mammalian target of rapamycin (mTOR)-dependent malformations of cortical development (MCDs) associated with seizures display hyperperfusion and increased vessel density of the dysmorphic cortical tissue. Some studies have suggested that the vascular defect occurred independently of seizures. Here, we further examined whether hypervascularization occurs in animal models of global and focal MCD with and without seizures, and whether it is sensitive to the mTOR blocker, rapamycin, that is approved for epilepsy treatment in tuberous sclerosis complex. METHODS: We used two experimental models of mTOR-dependent MCD consisting of conditional transgenic mice containing Tsc1null cells in the forebrain generating a global malformation associated with seizures and of wild-type mice containing a focal malformation in the somatosensory cortex generated by in utero electroporation (IUE) that does not lead to seizures. Alterations in blood vessels and the effects of a 2-week-long rapamycin treatment on these phenotypes were assessed in juvenile mice. RESULTS: Blood vessels in both the focal and global MCDs of postnatal day 14 mice displayed significant increase in vessel density, branching index, total vessel length, and decreased tissue lacunarity. In addition, rapamycin treatment (0.5 mg/kg, every 2 days) partially rescued vessel abnormalities in the focal MCD model, but it did not ameliorate the vessel abnormalities in the global MCD model that required higher rapamycin dosage for a partial rescue. SIGNIFICANCE: Here, we identified hypervascularization in mTOR-dependent MCD in the absence of seizures in young mice, suggesting that increased angiogenesis occurs during development in parallel to alterations in corticogenesis. In addition, a predictive functional outcome is that dysplastic neurons forming MCD will have better access to oxygen and metabolic supplies via their closer proximity to blood vessels. Finally, the difference in rapamycin sensitivity between a focal and global MCD suggest that rapamycin treatment will need to be titrated to match the type of MCD.


Asunto(s)
Malformaciones del Desarrollo Cortical/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Vasos Sanguíneos/patología , Tamaño de la Célula , Dendritas/patología , Electroporación , Femenino , Ratones , Ratones Transgénicos , Neovascularización Patológica/patología , Neuronas/patología , Plásmidos/genética , Embarazo , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Convulsiones/patología , Corteza Somatosensorial/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Esclerosis Tuberosa/complicaciones , Esclerosis Tuberosa/tratamiento farmacológico
12.
J Neurosci ; 39(14): 2762-2773, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30700531

RESUMEN

Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are focal malformations of cortical development (FMCDs) that are highly associated with intractable epilepsy. TSC and FCD are mTORopathies caused by a spectrum of pathogenic variants in the mechanistic target of rapamycin (mTOR) pathway genes leading to differential activation of mTOR signaling. However, whether the degree of mTOR hyperactivity influences disease severity remains unclear. Here, we examined the effects of differential mTOR hyperactivity levels on epilepsy and associated neuropathology in a mouse model of TSC and FCD. Constitutively active Rheb (RhebCA), the canonical activator of mTOR complex 1 (mTORC1), was expressed in mouse embryos of either sex via in utero electroporation at low, intermediate, and high concentrations to induce different mTORC1 activity levels in developing cortical neurons. We found that RhebCA expression induced mTORC1 hyperactivation and increased neuronal soma size and misplacement in a dose-dependent manner. No seizures were detected in the low RhebCA mice, whereas the intermediate and high RhebCA mice displayed spontaneous, recurrent seizures that significantly increased with higher RhebCA concentrations. Seizures were associated with a global increase in microglial activation that was notably higher in the regions containing RhebCA-expressing neurons. These data demonstrate that neuronal mTOR hyperactivity levels influence the severity of epilepsy and associated neuropathology in experimental TSC and FCD. Overall, these findings highlight the importance of evaluating the outcome of individual variants on mTOR activity levels and support personalized medicine strategies based on patient variants and mTOR activity level for TSC, FCD, and potentially other mTORopathies.SIGNIFICANCE STATEMENT Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are epileptogenic cortical malformations caused by pathogenic variants in mechanistic target of rapamycin (mTOR) pathway genes leading to differential mTOR hyperactivation. Here, we present novel findings that neuronal mTOR hyperactivity levels correlate with the severity of epilepsy and associated neuropathology in a mouse model of TSC and FCD. Our findings suggest the need to evaluate the outcome of individual variants on mTOR activity levels in clinical assessments and support personalized medicine strategies based on patient variants and mTOR activity level. Additionally, we present useful modifications to a previously described mouse model of TSC and FCD that allows for titration of seizure frequency and generation of a mild to severe epilepsy phenotype as applicable for preclinical drug testing and mechanistic studies.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/metabolismo , Malformaciones del Desarrollo Cortical/metabolismo , Índice de Severidad de la Enfermedad , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Electroencefalografía/métodos , Epilepsia/fisiopatología , Femenino , Masculino , Malformaciones del Desarrollo Cortical/fisiopatología , Ratones , Esclerosis Tuberosa/fisiopatología
13.
Neurosci Res ; 143: 44-52, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29857015

RESUMEN

We combined translating ribosome affinity purification (TRAP) with in utero electroporation (IUE), called iTRAP to identify the molecular profile of specific neuronal populations during neonatal development without the need for viral approaches and FACS sorting. We electroporated a plasmid encoding EGFP-tagged ribosomal protein L10a at embryonic day (E) 14-15 to target layer 2-4 cortical neurons of the somatosensory cortex. At three postnatal (P) ages-P0, P7, and P14-when morphogenesis occurs and synapses are forming, TRAP and molecular profiling was performed from electroporated regions. We found that ribosome bound (Ribo)-mRNAs from ∼7300 genes were significantly altered over time and included classical neuronal genes known to decrease (e.g., Tbr1, Dcx) or increase (e.g., Eno2, Camk2a, Syn1) as neurons mature. This approach led to the identification of specific developmental patterns for Ribo-mRNAs not previously reported to be developmentally regulated in neurons, providing rationale for future examination of their role in selective biological processes. These include upregulation of Lynx1, Nrn1, Cntnap1 over time; downregulation of St8sia2 and Draxin; and bidirectional changes to Fkbp1b. iTRAP is a versatile approach that allows researchers to easily assess the molecular profile of specific neuronal populations in selective brain regions under various conditions, including overexpression and knockdown of target genes, and in disease settings.


Asunto(s)
Electroporación/métodos , Biosíntesis de Proteínas , Células Piramidales/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Proteína Doblecortina , Desarrollo Embrionario , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Células Piramidales/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína Ribosómica L10 , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Corteza Somatosensorial/citología , Corteza Somatosensorial/metabolismo , Sinapsinas/metabolismo
14.
Nat Commun ; 7: 11753, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27249187

RESUMEN

Focal cortical dysplasia (FCD), a local malformation of cortical development, is the most common cause of pharmacoresistant epilepsy associated with life-long neurocognitive impairments. It remains unclear whether neuronal misplacement is required for seizure activity. Here we show that dyslamination and white matter heterotopia are not necessary for seizure generation in a murine model of type II FCDs. These experimental FCDs generated by increasing mTOR activity in layer 2/3 neurons of the medial prefrontal cortex are associated with tonic-clonic seizures and a normal survival rate. Preventing all FCD-related defects, including neuronal misplacement and dysmorphogenesis, with rapamycin treatments from birth eliminates seizures, but seizures recur after rapamycin withdrawal. In addition, bypassing neuronal misplacement and heterotopia using inducible vectors do not prevent seizure occurrence. Collectively, data obtained using our new experimental FCD-associated epilepsy suggest that life-long treatment to reduce neuronal dysmorphogenesis is required to suppress seizures in individuals with FCD.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Malformaciones del Desarrollo Cortical/tratamiento farmacológico , Neuronas/metabolismo , Convulsiones/tratamiento farmacológico , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Animales , Movimiento Celular , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical/patología , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
15.
Neurosci Lett ; 629: 15-18, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27345385

RESUMEN

Most individuals with tuberous sclerosis complex (TSC) are born with a mutant allele of either TSC1 or TSC2 and a mosaic of psychological and cognitive defects. Tsc1 loss of heterozygosity contributes to severe dendritic abnormalities that are rescued by normalizing the levels of the actin-cross linking protein, Filamin A (FLNA). However, it is unclear whether dendrites and FLNA levels are abnormal in an heterozygote Tsc1 condition. Here, we examined dendritic morphology and FLNA levels in the olfactory bulb of Tsc1 wild type and heterozygote mice. Using in vivo neonatal electroporation to label newborn neurons followed by sholl analysis, we found that Tsc1 haploinsufficiency is associated with increased dendritic complexity and total dendritic length as well as increased FLNA levels. Since reducing FLNA levels has been shown to decrease Tsc1(+/-) dendritic complexity, these data suggest that increased FLNA levels in Tsc1(+/-) mice contribute to abnormal dendritic patterning in the Tsc1 heterozygote condition of individuals with TSC.


Asunto(s)
Dendritas/genética , Dendritas/patología , Filaminas/metabolismo , Esclerosis Tuberosa/genética , Proteínas Supresoras de Tumor/genética , Animales , Haploinsuficiencia , Ratones , Ratones Transgénicos , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa
16.
Nat Neurosci ; 19(7): 905-14, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27239938

RESUMEN

Developmental nicotine exposure causes persistent changes in cortical neuron morphology and in behavior. We used microarray screening to identify master transcriptional or epigenetic regulators mediating these effects of nicotine and discovered increases in Ash2l mRNA, encoding a component of a histone methyltransferase complex. We therefore examined genome-wide changes in trimethylation of histone H3 on Lys4 (H3K4me3), a mark induced by the Ash2l complex associated with increased gene transcription. A large proportion of regulated promoter sites were involved in synapse maintenance. We found that Mef2c interacts with Ash2l and mediates changes in H3K4me3. Knockdown of Ash2l or Mef2c abolished nicotine-mediated alterations of dendritic complexity in vitro and in vivo, and attenuated nicotine-dependent changes in passive avoidance behavior. In contrast, overexpression mimicked nicotine-mediated alterations of neuronal structure and passive avoidance behavior. These studies identify Ash2l as a target induced by nicotinic stimulation that couples developmental nicotine exposure to changes in brain epigenetic marks, neuronal structure and behavior.


Asunto(s)
Proteínas de Unión al ADN/genética , Epigénesis Genética/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , Nicotina/farmacología , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Animales , Histonas/metabolismo , Metilación/efectos de los fármacos , Ratones Endogámicos C57BL
17.
Neurosci Lett ; 612: 43-47, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26655465

RESUMEN

Expression of hypoxia-inducible factor 1a (HIF1a) is increased under several pathological conditions such as hyperactive mechanistic target of rapamycin complex 1 (mTORC1) in tuberous sclerosis complex (TSC). Hyperactive mTORC1 and the resulting increased dendritic complexity of neurons are shared molecular and cellular alterations in several neurological disorders associated with cognitive disabilities. Despite some evidence that HIF1a contributes to dendritic overgrowth in vitro, it remains unknown whether increased HIF1a in TSC neurons could contribute to their increased dendritic complexity. To address this use in vivo, we generated TSC neurons by deleting Tsc1 in newborn olfactory bulb (OB) neurons of conditional Tsc1 transgenic mice using neonatal electroporation. In addition to their increased dendritic complexity, Tsc1(null) neurons have been reported to display increased Hif1a mRNA level and HIF1a transcriptional activity. We found that Tsc1(null)-dependent dendritic overgrowth was prevented by knocking down HIF1a or expressing a dominant negative HIF1a. In addition, overexpressing HIF1a in wild-type developing neurons resulted in increased dendritic complexity in vivo. These data highlight that an increase in HIF1a levels contributes to abnormal dendritic patterning in developing neurons under normal conditions and hyperactive mTORC1 conditions as in TSC.


Asunto(s)
Dendritas/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Esclerosis Tuberosa/metabolismo , Animales , Animales Recién Nacidos , Femenino , Técnicas de Silenciamiento del Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones Transgénicos , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
18.
Neuron ; 84(1): 78-91, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25277454

RESUMEN

Abnormal dendritic complexity is a shared feature of many neurodevelopmental disorders associated with neurological defects. Here, we found that the actin-crosslinking protein filamin A (FLNA) is overexpressed in tuberous sclerosis complex (TSC) mice, a PI3K-mTOR model of neurodevelopmental disease that is associated with abnormal dendritic complexity. Both under- and overexpression of FLNA in wild-type neurons led to more complex dendritic arbors in vivo, suggesting that an optimal level of FLNA expression is required for normal dendritogenesis. In Tsc1(null) neurons, knocking down FLNA in vivo prevented dendritic abnormalities. Surprisingly, FLNA overexpression in Tsc1(null) neurons was dependent on MEK1/2 but not mTOR activity, despite both pathways being hyperactive. In addition, increasing MEK-ERK1/2 activity led to dendritic abnormalities via FLNA, and decreasing MEK-ERK1/2 signaling in Tsc1(null) neurons rescued dendritic defects. These data demonstrate that altered FLNA expression increases dendritic complexity and contributes to pathologic dendritic patterning in TSC in an mTOR-independent, ERK1/2-dependent manner.


Asunto(s)
Dendritas/metabolismo , Filaminas/biosíntesis , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/metabolismo , Adulto , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Dendritas/patología , Femenino , Filaminas/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Técnicas de Cultivo de Órganos , Esclerosis Tuberosa/patología , Adulto Joven
19.
PLoS One ; 9(5): e96956, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24806451

RESUMEN

Hyperactive mammalian target of rapamycin (mTOR) is associated with cognitive deficits in several neurological disorders including tuberous sclerosis complex (TSC). The phosphorylation of the mRNA-binding protein FMRP reportedly depends on mTOR complex 1 (mTORC1) activity via p70 S6 kinase 1 (S6K1). Because this phosphorylation is thought to regulate the translation of messages important for synaptic plasticity, we explored whether FMRP phosphorylation of the S6K1-dependent residue (S499) is altered in TSC and states of dysregulated TSC-mTORC1 signaling. Surprisingly, we found that FMRP S499 phosphorylation was unchanged in heterozygous and conditional Tsc1 knockout mice despite significantly elevated mTORC1-S6K1 activity. Neither up- nor down-regulation of the mTORC1-S6K1 axis in vivo or in vitro had any effect on phospho-FMRP S499 levels. In addition, FMRP S499 phosphorylation was unaltered in S6K1-knockout mice. Collectively, these data strongly suggest that FMRP S499 phosphorylation is independent of mTORC1-S6K1 activity and is not altered in TSC.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Esclerosis Tuberosa/genética , Animales , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Plasticidad Neuronal/genética , Fosforilación , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Esclerosis Tuberosa/mortalidad , Esclerosis Tuberosa/patología
20.
Ann Thorac Surg ; 97(3): 879-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24206970

RESUMEN

BACKGROUND: Numerous gaseous microemboli (GME) are delivered into the arterial circulation during cardiopulmonary bypass (CPB). These emboli damage end organs through multiple mechanisms that are thought to contribute to neurocognitive deficits after cardiac surgery. Here, we use hypobaric oxygenation to reduce dissolved gases in blood and greatly reduce GME delivery during CPB. METHODS: Variable subatmospheric pressures were applied to 100% oxygen sweep gas in standard hollow fiber microporous membrane oxygenators to oxygenate and denitrogenate blood. GME were quantified using ultrasound while air embolism from the surgical field was simulated experimentally. We assessed end-organ tissues in swine postoperatively using light microscopy. RESULTS: Variable sweep gas pressures allowed reliable oxygenation independent of carbon dioxide removal while denitrogenating arterial blood. Hypobaric oxygenation produced dose-dependent reductions of Doppler signals produced by bolus and continuous GME loads in vitro. Swine were maintained using hypobaric oxygenation for 4 hours on CPB with no apparent adverse events. Compared with current practice standards of oxygen/air sweep gas, hypobaric oxygenation reduced GME volumes exiting the oxygenator (by 80%), exiting the arterial filter (95%), and arriving at the aortic cannula (∼100%), indicating progressive reabsorption of emboli throughout the CPB circuit in vivo. Analysis of brain tissue suggested decreased microvascular injury under hypobaric conditions. CONCLUSIONS: Hypobaric oxygenation is an effective, low-cost, common sense approach that capitalizes on the simple physical makeup of GME to achieve their near-total elimination during CPB. This technique holds great potential for limiting end-organ damage and improving outcomes in a variety of patients undergoing extracorporeal circulation.


Asunto(s)
Puente Cardiopulmonar/efectos adversos , Embolia Aérea/etiología , Embolia Aérea/terapia , Terapia por Inhalación de Oxígeno/métodos , Animales , Embolia Aérea/patología , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA