Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Nat Med ; 30(4): 1096-1103, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38622249

RESUMO

Prasinezumab, a monoclonal antibody that binds aggregated α-synuclein, is being investigated as a potential disease-modifying therapy in early-stage Parkinson's disease. Although in the PASADENA phase 2 study, the primary endpoint (Movement Disorder Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) sum of Parts I + II + III) was not met, prasinezumab-treated individuals exhibited slower progression of motor signs than placebo-treated participants (MDS-UPDRS Part III). We report here an exploratory analysis assessing whether prasinezumab showed greater benefits on motor signs progression in prespecified subgroups with faster motor progression. Prasinezumab's potential effects on disease progression were assessed in four prespecified and six exploratory subpopulations of PASADENA: use of monoamine oxidase B inhibitors at baseline (yes versus no); Hoehn and Yahr stage (2 versus 1); rapid eye movement sleep behavior disorder (yes versus no); data-driven subphenotypes (diffuse malignant versus nondiffuse malignant); age at baseline (≥60 years versus <60 years); sex (male versus female); disease duration (>12 months versus <12 months); age at diagnosis (≥60 years versus <60 years); motor subphenotypes (akinetic-rigid versus tremor-dominant); and motor subphenotypes (postural instability gait dysfunction versus tremor-dominant). In these subpopulations, the effect of prasinezumab on slowing motor signs progression (MDS-UPDRS Part III) was greater in the rapidly progressing subpopulations (for example, participants who were diffuse malignant or taking monoamine oxidase B inhibitors at baseline). This exploratory analysis suggests that, in a trial of 1-year duration, prasinezumab might reduce motor progression to a greater extent in individuals with more rapidly progressing Parkinson's disease. However, because this was a post hoc analysis, additional randomized clinical trials are needed to validate these findings.


Assuntos
Doença de Parkinson , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Tremor/tratamento farmacológico , Antiparkinsonianos/uso terapêutico , Monoaminoxidase/uso terapêutico , Progressão da Doença
2.
bioRxiv ; 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-37961719

RESUMO

Precise control of protein ubiquitination is essential for brain development, and hence, disruption of ubiquitin signaling networks can lead to neurological disorders. Mutations of the deubiquitinase USP7 cause the Hao-Fountain syndrome (HAFOUS), characterized by developmental delay, intellectual disability, autism, and aggressive behavior. Here, we report that conditional deletion of USP7 in excitatory neurons in the mouse forebrain triggers diverse phenotypes including sensorimotor deficits, learning and memory impairment, and aggressive behavior, resembling clinical features of HAFOUS. USP7 deletion induces neuronal apoptosis in a manner dependent of the tumor suppressor p53. However, most behavioral abnormalities in USP7 conditional mice persist despite p53 loss. Strikingly, USP7 deletion in the brain perturbs the synaptic proteome and dendritic spine morphogenesis independently of p53. Integrated proteomics analysis reveals that the neuronal USP7 interactome is enriched for proteins implicated in neurodevelopmental disorders and specifically identifies the RNA splicing factor Ppil4 as a novel neuronal substrate of USP7. Knockdown of Ppil4 in cortical neurons impairs dendritic spine morphogenesis, phenocopying the effect of USP7 loss on dendritic spines. These findings reveal a novel USP7-Ppil4 ubiquitin signaling link that regulates neuronal connectivity in the developing brain, with implications for our understanding of the pathogenesis of HAFOUS and other neurodevelopmental disorders.

3.
PLoS One ; 18(11): e0294731, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38015828

RESUMO

Astrocyte-specific ion pump α2-Na+/K+-ATPase plays a critical role in the pathogenesis of amyotrophic lateral sclerosis (ALS). Here, we test the effect of Atp1a2 mRNA-specific antisense oligonucleotides (ASOs) to induce α2-Na+/K+-ATPase knockdown in the widely used ALS animal model, SOD1*G93A mice. Two ASOs led to efficient Atp1a2 knockdown and significantly reduced SOD1 aggregation in vivo. Although Atp1a2 ASO-treated mice displayed no off-target or systemic toxicity, the ASO-treated mice exhibited an accelerated disease onset and shorter lifespan than control mice. Transcriptomics studies reveal downregulation of genes involved in oxidative response, metabolic pathways, trans-synaptic signaling, and upregulation of genes involved in glutamate receptor signaling and complement activation, suggesting a potential role for these molecular pathways in de-coupling SOD1 aggregation from survival in Atp1a2 ASO-treated mice. Together, these results reveal a role for α2-Na+/K+-ATPase in SOD1 aggregation and highlight the critical effect of temporal modulation of genetically validated therapeutic targets in neurodegenerative diseases.


Assuntos
Esclerose Lateral Amiotrófica , Animais , Camundongos , Esclerose Lateral Amiotrófica/patologia , Astrócitos/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Neurônios Motores/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Medula Espinal/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
4.
Cell Death Dis ; 14(7): 405, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37414747

RESUMO

Protein post-translational modification by the small ubiquitin-like modifier (SUMO) regulates the stability, subcellular localization, and interactions of protein substrates with consequences on cellular responses including epithelial-mesenchymal transition (EMT). Transforming growth factor beta (TGFß) is a potent inducer of EMT with implications for cancer invasion and metastasis. The transcriptional coregulator SnoN suppresses TGFß-induced EMT-associated responses in a sumoylation-dependent manner, but the underlying mechanisms have remained largely unknown. Here, we find that sumoylation promotes the interaction of SnoN with the epigenetic regulators histone deacetylase 1 (HDAC1) and histone acetylase p300 in epithelial cells. In gain and loss of function studies, HDAC1 suppresses, whereas p300 promotes, TGFß-induced morphogenetic changes associated with EMT-related events in three-dimensional multicellular organoids derived from mammary epithelial cells or carcinomas. These findings suggest that sumoylated SnoN acts via the regulation of histone acetylation to modulate EMT-related effects in breast cell organoids. Our study may facilitate the discovery of new biomarkers and therapeutics in breast cancer and other epithelial cell-derived cancers.


Assuntos
Transição Epitelial-Mesenquimal , Histona Desacetilase 1 , Histona Desacetilase 1/genética , Organoides/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fenótipo
5.
Cell Death Differ ; 28(1): 267-282, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32770107

RESUMO

SUMO E3 ligases specify protein substrates for SUMOylation. The SUMO E3 ligases PIAS1 and TIF1γ target the transcriptional regulator SnoN for SUMOylation leading to suppression of epithelial-mesenchymal transition (EMT). Whether and how TIF1γ and PIAS1 might coordinate SnoN SUMOylation and regulation of EMT remained unknown. Here, we reveal that SnoN associates simultaneously with both TIF1γ and PIAS1, leading to a trimeric protein complex. Hence, PIAS1 and TIF1γ collaborate to promote the SUMOylation of SnoN. Importantly, loss of function studies of PIAS1 and TIF1γ suggest that these E3 ligases act in an interdependent manner to suppress EMT of breast cell-derived tissue organoids. Collectively, our findings unveil a novel mechanism by which SUMO E3 ligases coordinate substrate SUMOylation with biological implications.


Assuntos
Transição Epitelial-Mesenquimal/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Inibidoras de STAT Ativados/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Sumoilação/genética , Fatores de Transcrição/genética , Animais , Técnicas de Cultura de Células em Três Dimensões , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
6.
Nat Commun ; 11(1): 6164, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33268780

RESUMO

Familial hemiplegic migraine is an episodic neurological disorder characterized by transient sensory and motor symptoms and signs. Mutations of the ion pump α2-Na/K ATPase cause familial hemiplegic migraine, but the mechanisms by which α2-Na/K ATPase mutations lead to the migraine phenotype remain incompletely understood. Here, we show that mice in which α2-Na/K ATPase is conditionally deleted in astrocytes display episodic paralysis. Functional neuroimaging reveals that conditional α2-Na/K ATPase knockout triggers spontaneous cortical spreading depression events that are associated with EEG low voltage activity events, which correlate with transient motor impairment in these mice. Transcriptomic and metabolomic analyses show that α2-Na/K ATPase loss alters metabolic gene expression with consequent serine and glycine elevation in the brain. A serine- and glycine-free diet rescues the transient motor impairment in conditional α2-Na/K ATPase knockout mice. Together, our findings define a metabolic mechanism regulated by astrocytic α2-Na/K ATPase that triggers episodic motor paralysis in mice.


Assuntos
Astrócitos/metabolismo , Ataxia/genética , Metaboloma/genética , Enxaqueca com Aura/genética , ATPase Trocadora de Sódio-Potássio/genética , Transcriptoma , Animais , Astrócitos/patologia , Ataxia/metabolismo , Ataxia/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Neuroimagem Funcional , Glicina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Enxaqueca com Aura/metabolismo , Enxaqueca com Aura/patologia , Teste de Desempenho do Rota-Rod , Serina/metabolismo , ATPase Trocadora de Sódio-Potássio/deficiência
7.
Cell Death Dis ; 11(8): 704, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843642

RESUMO

Alternative splicing contributes to diversification of gene function, yet consequences of splicing on functions of specific gene products is poorly understood. The major transcription factor TCF7L2 undergoes alternative splicing but the biological significance of TCF7L2 isoforms has remained largely to be elucidated. Here, we find that the TCF7L2 E-isoforms maintain, whereas the M and S isoforms disrupt morphogenesis of 3D-epithelial cell-derived organoids via regulation of epithelial-mesenchymal transition (EMT). Remarkably, TCF7L2E2 antagonizes, whereas TCF7L2M2/S2 promotes EMT-like effects in epithelial cells induced by transforming growth factor beta (TGFß) signaling. In addition, we find TGFß signaling reduces the proportion of TCF7L2E to TCF7L2M/S protein in cells undergoing EMT. We also find that TCF7L2 operates via TGFß-Smad3 signaling to regulate EMT. Collectively, our findings unveil novel isoform-specific functions for the major transcription factor TCF7L2 and provide novel links between TCF7L2 and TGFß signaling in the control of EMT-like responses and epithelial tissue morphogenesis.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Organoides/fisiologia , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Humanos , Camundongos , Morfogênese/efeitos dos fármacos , Isoformas de Proteínas , Transdução de Sinais/fisiologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
8.
BMC Bioinformatics ; 21(1): 269, 2020 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-32600248

RESUMO

BACKGROUND: High throughput RNA sequencing is a powerful approach to study gene expression. Due to the complex multiple-steps protocols in data acquisition, extreme deviation of a sample from samples of the same treatment group may occur due to technical variation or true biological differences. The high-dimensionality of the data with few biological replicates make it challenging to accurately detect those samples, and this issue is not well studied in the literature currently. Robust statistics is a family of theories and techniques aim to detect the outliers by first fitting the majority of the data and then flagging data points that deviate from it. Robust statistics have been widely used in multivariate data analysis for outlier detection in chemometrics and engineering. Here we apply robust statistics on RNA-seq data analysis. RESULTS: We report the use of two robust principal component analysis (rPCA) methods, PcaHubert and PcaGrid, to detect outlier samples in multiple simulated and real biological RNA-seq data sets with positive control outlier samples. PcaGrid achieved 100% sensitivity and 100% specificity in all the tests using positive control outliers with varying degrees of divergence. We applied rPCA methods and classical principal component analysis (cPCA) on an RNA-Seq data set profiling gene expression of the external granule layer in the cerebellum of control and conditional SnoN knockout mice. Both rPCA methods detected the same two outlier samples but cPCA failed to detect any. We performed differentially expressed gene detection before and after outlier removal as well as with and without batch effect modeling. We validated gene expression changes using quantitative reverse transcription PCR and used the result as reference to compare the performance of eight different data analysis strategies. Removing outliers without batch effect modeling performed the best in term of detecting biologically relevant differentially expressed genes. CONCLUSIONS: rPCA implemented in the PcaGrid function is an accurate and objective method to detect outlier samples. It is well suited for high-dimensional data with small sample sizes like RNA-seq data. Outlier removal can significantly improve the performance of differential gene detection and downstream functional analysis.


Assuntos
Análise de Componente Principal , RNA-Seq/métodos , Animais , Cerebelo/metabolismo , Feminino , Masculino , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Neurosci ; 39(1): 44-62, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30425119

RESUMO

Control of neuronal precursor cell proliferation is essential for normal brain development, and deregulation of this fundamental developmental event contributes to brain diseases. Typically, neuronal precursor cell proliferation extends over long periods of time during brain development. However, how neuronal precursor proliferation is regulated in a temporally specific manner remains to be elucidated. Here, we report that conditional KO of the transcriptional regulator SnoN in cerebellar granule neuron precursors robustly inhibits the proliferation of these cells and promotes their cell cycle exit at later stages of cerebellar development in the postnatal male and female mouse brain. In laser capture microdissection followed by RNA-Seq, designed to profile gene expression specifically in the external granule layer of the cerebellum, we find that SnoN promotes the expression of cell proliferation genes and concomitantly represses differentiation genes in granule neuron precursors in vivo Remarkably, bioinformatics analyses reveal that SnoN-regulated genes contain binding sites for the transcription factors N-myc and Pax6, which promote the proliferation and differentiation of granule neuron precursors, respectively. Accordingly, we uncover novel physical interactions of SnoN with N-myc and Pax6 in cells. In behavior analyses, conditional KO of SnoN impairs cerebellar-dependent learning in a delayed eye-blink conditioning paradigm, suggesting that SnoN-regulation of granule neuron precursor proliferation bears functional consequences at the organismal level. Our findings define a novel function and mechanism for the major transcriptional regulator SnoN in the control of granule neuron precursor proliferation in the mammalian brain.SIGNIFICANCE STATEMENT This study reports the discovery that the transcriptional regulator SnoN plays a crucial role in the proliferation of cerebellar granule neuron precursors in the postnatal mouse brain. Conditional KO of SnoN in granule neuron precursors robustly inhibits the proliferation of these cells and promotes their cycle exit specifically at later stages of cerebellar development, with biological consequences of impaired cerebellar-dependent learning. Genomics and bioinformatics analyses reveal that SnoN promotes the expression of cell proliferation genes and concomitantly represses cell differentiation genes in vivo Although SnoN has been implicated in distinct aspects of the development of postmitotic neurons, this study identifies a novel function for SnoN in neuronal precursors in the mammalian brain.


Assuntos
Encéfalo/citologia , Proliferação de Células , Cerebelo/fisiologia , Células-Tronco Neurais/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Animais , Comportamento Animal , Piscadela/fisiologia , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular/genética , Cerebelo/citologia , Biologia Computacional , Grânulos Citoplasmáticos/fisiologia , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Genes myc/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/fisiologia
10.
Mol Autism ; 10: 51, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31893020

RESUMO

Background: Autism spectrum disorder (ASD) is a neurodevelopmental disorder with pronounced heritability in the general population. This is largely attributable to the effects of polygenic susceptibility, with inherited liability exhibiting distinct sex differences in phenotypic expression. Attempts to model ASD in human cellular systems have principally involved rare de novo mutations associated with ASD phenocopies. However, by definition, these models are not representative of polygenic liability, which accounts for the vast share of population-attributable risk. Methods: Here, we performed what is, to our knowledge, the first attempt to model multiplex autism using patient-derived induced pluripotent stem cells (iPSCs) in a family manifesting incremental degrees of phenotypic expression of inherited liability (absent, intermediate, severe). The family members share an inherited variant of uncertain significance (VUS) in GPD2, a gene that was previously associated with developmental disability but here is insufficient by itself to cause ASD. iPSCs from three first-degree relatives and an unrelated control were differentiated into both cortical excitatory (cExN) and cortical inhibitory (cIN) neurons, and cellular phenotyping and transcriptomic analysis were conducted. Results: cExN neurospheres from the two affected individuals were reduced in size, compared to those derived from unaffected related and unrelated individuals. This reduction was, at least in part, due to increased apoptosis of cells from affected individuals upon initiation of cExN neural induction. Likewise, cIN neural progenitor cells from affected individuals exhibited increased apoptosis, compared to both unaffected individuals. Transcriptomic analysis of both cExN and cIN neural progenitor cells revealed distinct molecular signatures associated with affectation, including the misregulation of suites of genes associated with neural development, neuronal function, and behavior, as well as altered expression of ASD risk-associated genes. Conclusions: We have provided evidence of morphological, physiological, and transcriptomic signatures of polygenic liability to ASD from an analysis of cellular models derived from a multiplex autism family. ASD is commonly inherited on the basis of additive genetic liability. Therefore, identifying convergent cellular and molecular phenotypes resulting from polygenic and monogenic susceptibility may provide a critical bridge for determining which of the disparate effects of rare highly deleterious mutations might also apply to common autistic syndromes.


Assuntos
Transtorno Autístico/patologia , Comunicação Celular , Células-Tronco Pluripotentes Induzidas/patologia , Neurônios/patologia , Adolescente , Transtorno Autístico/genética , Diferenciação Celular/genética , Pré-Escolar , Análise por Conglomerados , Família , Feminino , Regulação da Expressão Gênica , Genótipo , Humanos , Lactente , Recém-Nascido , Interneurônios/patologia , Masculino , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Linhagem , Fenótipo , Gravidez , Reprodutibilidade dos Testes , Transcriptoma/genética
11.
Cell Rep ; 25(6): 1404-1414.e6, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30403997

RESUMO

Mutations of the transcriptional regulator PHF6 cause the X-linked intellectual disability disorder Börjeson-Forssman-Lehmann syndrome (BFLS), but the pathogenesis of BFLS remains poorly understood. Here, we report a mouse model of BFLS, generated using a CRISPR-Cas9 approach, in which cysteine 99 within the PHD domain of PHF6 is replaced with phenylalanine (C99F). Mice harboring the patient-specific C99F mutation display deficits in cognitive functions, emotionality, and social behavior, as well as reduced threshold to seizures. Electrophysiological studies reveal that the intrinsic excitability of entorhinal cortical stellate neurons is increased in PHF6 C99F mice. Transcriptomic analysis of the cerebral cortex in C99F knockin mice and PHF6 knockout mice show that PHF6 promotes the expression of neurogenic genes and represses synaptic genes. PHF6-regulated genes are also overrepresented in gene signatures and modules that are deregulated in neurodevelopmental disorders of cognition. Our findings advance our understanding of the mechanisms underlying BFLS pathogenesis.


Assuntos
Epilepsia/patologia , Face/anormalidades , Dedos/anormalidades , Transtornos do Crescimento/patologia , Hipogonadismo/patologia , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Obesidade/patologia , Animais , Sequência de Bases , Encéfalo/patologia , Proteínas de Transporte/genética , Cognição , Modelos Animais de Doenças , Suscetibilidade a Doenças , Emoções , Epilepsia/genética , Face/patologia , Dedos/patologia , Regulação da Expressão Gênica , Transtornos do Crescimento/genética , Hipogonadismo/genética , Relações Interpessoais , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/genética , Camundongos , Camundongos Mutantes , Neurônios/metabolismo , Neurônios/patologia , Obesidade/genética , Proteínas Repressoras , Convulsões/patologia , Sinapses/metabolismo , Transcrição Gênica
13.
Nat Neurosci ; 19(6): 798-806, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27110918

RESUMO

EGFRvIII-STAT3 signaling is important in glioblastoma pathogenesis. Here, we identified the cytokine receptor OSMR as a direct target gene of the transcription factor STAT3 in mouse astrocytes and human brain tumor stem cells (BTSCs). We found that OSMR functioned as an essential co-receptor for EGFRvIII. OSMR formed a physical complex with EGFRvIII, and depletion of OSMR impaired EGFRvIII-STAT3 signaling. Conversely, pharmacological inhibition of EGFRvIII phosphorylation inhibited the EGFRvIII-OSMR interaction and activation of STAT3. EGFRvIII-OSMR signaling in tumors operated constitutively, whereas EGFR-OSMR signaling in nontumor cells was synergistically activated by the ligands EGF and OSM. Finally, knockdown of OSMR strongly suppressed cell proliferation and tumor growth of mouse glioblastoma cells and human BTSC xenografts in mice, and prolonged the lifespan of these mice. Our findings identify OSMR as a critical regulator of glioblastoma tumor growth that orchestrates a feed-forward signaling mechanism with EGFRvIII and STAT3 to drive tumorigenesis.


Assuntos
Neoplasias Encefálicas/metabolismo , Transformação Celular Neoplásica/metabolismo , Citocinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Transdução de Sinais/fisiologia , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos Transgênicos , Transplante de Neoplasias/métodos , Fator de Transcrição STAT3/metabolismo
14.
Trends Neurosci ; 38(7): 439-47, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26100142

RESUMO

Dendrites play a central role in the integration and flow of information in the nervous system. The morphogenesis and maturation of dendrites is hence an essential step in the establishment of neuronal connectivity. Recent studies have uncovered crucial functions for extrinsic cues in the development of dendrites. We review the contribution of secreted polypeptide growth factors, contact-mediated proteins, and neuronal activity in distinct phases of dendrite development. We also highlight how extrinsic cues influence local and global intracellular mechanisms of dendrite morphogenesis. Finally, we discuss how these studies have advanced our understanding of neuronal connectivity and have shed light on the pathogenesis of neurodevelopmental disorders.


Assuntos
Sinais (Psicologia) , Dendritos/fisiologia , Morfogênese/fisiologia , Neurônios/citologia , Animais , Sinalização do Cálcio , Humanos , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurotransmissores/metabolismo
15.
Nat Neurosci ; 17(12): 1710-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25344630

RESUMO

Perturbations of astrocytes trigger neurodegeneration in several diseases, but the glial cell-intrinsic mechanisms that induce neurodegeneration remain poorly understood. We found that a protein complex of α2-Na/K ATPase and α-adducin was enriched in astrocytes expressing mutant superoxide dismutase 1 (SOD1), which causes familial amyotrophic lateral sclerosis (ALS). Knockdown of α2-Na/K ATPase or α-adducin in mutant SOD1 astrocytes protected motor neurons from degeneration, including in mutant SOD1 mice in vivo. Heterozygous disruption of the α2-Na/K ATPase gene suppressed degeneration in vivo and increased the lifespan of mutant SOD1 mice. The pharmacological agent digoxin, which inhibits Na/K ATPase activity, protected motor neurons from mutant SOD1 astrocyte-induced degeneration. Notably, α2-Na/K ATPase and α-adducin were upregulated in spinal cord of sporadic and familial ALS patients. Collectively, our findings define chronic activation of the α2-Na/K ATPase/α-adducin complex as a critical glial cell-intrinsic mechanism of non-cell autonomous neurodegeneration, with implications for potential therapies for neurodegenerative diseases.


Assuntos
Astrócitos/metabolismo , Astrócitos/patologia , Proteínas do Citoesqueleto/biossíntese , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , ATPase Trocadora de Sódio-Potássio/biossíntese , Animais , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
J Biol Chem ; 289(36): 25067-78, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25059663

RESUMO

Epithelial-mesenchymal transition (EMT) is a fundamental cellular process that contributes to epithelial tissue morphogenesis during normal development and in tumor invasiveness and metastasis. The transcriptional regulator SnoN robustly influences EMT in response to the cytokine TGFß, but the mechanisms that regulate the fundamental role of SnoN in TGFß-induced EMT are not completely understood. Here we employ interaction proteomics to uncover the signaling protein TIF1γ as a specific interactor of SnoN1 but not the closely related isoform SnoN2. A 16-amino acid peptide within a unique region of SnoN1 mediates the interaction of SnoN1 with TIF1γ. Strikingly, although TIF1γ is thought to act as a ubiquitin E3 ligase, we find that TIF1γ operates as a small ubiquitin-like modifier (SUMO) E3 ligase that promotes the sumoylation of SnoN1 at distinct lysine residues. Importantly, TIF1γ-induced sumoylation is required for the ability of SnoN1 to suppress TGFß-induced EMT, as assayed by the disruption of the morphogenesis of acini in a physiologically relevant three-dimensional model of normal murine mammary gland (NMuMG) epithelial cells. Collectively, our findings define a novel TIF1γ-SnoN1 sumoylation pathway that plays a critical role in EMT and has important implications for our understanding of TGFß signaling and diverse biological processes in normal development and cancer biology.


Assuntos
Transição Epitelial-Mesenquimal/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Western Blotting , Técnicas de Cultura de Células , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Sumoilação/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Ubiquitina-Proteína Ligases/metabolismo
17.
Neuron ; 83(1): 122-34, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24991957

RESUMO

Precise control of gene expression plays fundamental roles in brain development, but the roles of chromatin regulators in neuronal connectivity have remained poorly understood. We report that depletion of the NuRD complex by in vivo RNAi and conditional knockout of the core NuRD subunit Chd4 profoundly impairs the establishment of granule neuron parallel fiber/Purkinje cell synapses in the rodent cerebellar cortex in vivo. By interfacing genome-wide sequencing of transcripts and ChIP-seq analyses, we uncover a network of repressed genes and distinct histone modifications at target gene promoters that are developmentally regulated by the NuRD complex in the cerebellum in vivo. Finally, in a targeted in vivo RNAi screen of NuRD target genes, we identify a program of NuRD-repressed genes that operate as critical regulators of presynaptic differentiation in the cerebellar cortex. Our findings define NuRD-dependent promoter decommissioning as a developmentally regulated programming mechanism that drives synaptic connectivity in the mammalian brain.


Assuntos
Química Encefálica/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Regiões Promotoras Genéticas/genética , Sinapses/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células de Purkinje/fisiologia , Células de Purkinje/ultraestrutura , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Proteína 4 de Ligação ao Retinoblastoma/genética , Sinapses/ultraestrutura
18.
J Neurosci ; 33(46): 18015-21, 2013 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-24227713

RESUMO

Tuberous sclerosis complex (TSC) is a leading genetic cause of autism. The TSC proteins Tsc1 and Tsc2 control the mTORC1 signaling pathway in diverse cells, but how the mTORC1 pathway is specifically regulated in neurons remains to be elucidated. Here, using an interaction proteomics approach in neural cells including neurons, we uncover the brain-enriched protein huntingtin-associated protein 1 (Hap1) as a novel functional partner of Tsc1. Knockdown of Hap1 promotes specification of supernumerary axons in primary hippocampal neurons and profoundly impairs the positioning of pyramidal neurons in the mouse hippocampus in vivo. The Hap1 knockdown-induced phenotypes in primary neurons and in vivo recapitulate the phenotypes induced by Tsc1 knockdown. We also find that Hap1 knockdown in hippocampal neurons induces the downregulation of Tsc1 and stimulates the activity of mTORC1, as reflected by phosphorylation of the ribosomal protein S6. Inhibition of mTORC1 activity suppresses the Hap1 knockdown-induced polarity phenotype in hippocampal neurons. Collectively, these findings define a novel link between Hap1 and Tsc1 that regulates neuronal mTORC1 signaling and neuronal morphogenesis, with implications for our understanding of developmental disorders of cognition.


Assuntos
Encéfalo/crescimento & desenvolvimento , Morfogênese/fisiologia , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Ligação Proteica/fisiologia , Ratos , Transdução de Sinais/fisiologia , Proteína 1 do Complexo Esclerose Tuberosa
19.
PLoS One ; 8(6): e67178, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840619

RESUMO

Transforming growth factor-beta (TGFß) is a secreted polypeptide that plays essential roles in cellular development and homeostasis. Although mechanisms of TGFß-induced responses have been characterized, our understanding of TGFß signaling remains incomplete. Here, we uncover a novel function for the protein kinase NDR1 (nuclear Dbf2-related 1) in TGFß responses. Using an immunopurification approach, we find that NDR1 associates with SnoN, a key component of TGFß signaling. Knockdown of NDR1 by RNA interference promotes the ability of TGFß to induce transcription and cell cycle arrest in NMuMG mammary epithelial cells. Conversely, expression of NDR1 represses TGFß-induced transcription and inhibits the ability of TGFß to induce cell cycle arrest in NMuMG cells. Mechanistically, we find that NDR1 acts in a kinase-dependent manner to suppress the ability of TGFß to induce the phosphorylation and consequent nuclear accumulation of Smad2, which is critical for TGFß-induced transcription and responses. Strikingly, we also find that TGFß reciprocally regulates NDR1, whereby TGFß triggers the degradation of NDR1 protein. Collectively, our findings define a novel and intimate link between the protein kinase NDR1 and TGFß signaling. NDR1 suppresses TGFß-induced transcription and cell cycle arrest, and counteracting NDR1's negative regulation, TGFß signaling induces the downregulation of NDR1 protein. These findings advance our understanding of TGFß signaling, with important implications in development and tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Células Epiteliais/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Proliferação de Células , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Fosforilação , Proteólise , Proteínas Proto-Oncogênicas/metabolismo , Proteína Smad2/metabolismo , Transcrição Gênica
20.
FEBS Lett ; 586(14): 1977-83, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22710173

RESUMO

The transcriptional regulator SnoN plays a fundamental role as a modulator of transforming growth factor beta (TGFß)-induced signal transduction and biological responses. In recent years, novel functions of SnoN have been discovered in both TGFß-dependent and TGFß-independent settings in proliferating cells and postmitotic neurons. Accumulating evidence suggests that SnoN plays a dual role as a corepressor or coactivator of TGFß-induced transcription. Accordingly, SnoN exerts oncogenic or tumor-suppressive effects in epithelial tissues. At the cellular level, SnoN antagonizes or mediates the ability of TGFß to induce cell cycle arrest in a cell-type specific manner. SnoN also exerts key effects on epithelial-mesenchymal transition (EMT), with implications in cancer biology. Recent studies have expanded SnoN functions to postmitotic neurons, where SnoN orchestrates key aspects of neuronal development in the mammalian brain, from axon growth and branching to neuronal migration and positioning. In this review, we will highlight our understanding of SnoN biology at the crossroads of cancer biology and neurobiology.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Axônios/metabolismo , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitose , Modelos Biológicos , Modelos Neurológicos , Neoplasias/metabolismo , Neurônios/metabolismo , Isoformas de Proteínas , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA