Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 173(3): 792-803.e19, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29656897

RESUMO

Microscopy is a central method in life sciences. Many popular methods, such as antibody labeling, are used to add physical fluorescent labels to specific cellular constituents. However, these approaches have significant drawbacks, including inconsistency; limitations in the number of simultaneous labels because of spectral overlap; and necessary perturbations of the experiment, such as fixing the cells, to generate the measurement. Here, we show that a computational machine-learning approach, which we call "in silico labeling" (ISL), reliably predicts some fluorescent labels from transmitted-light images of unlabeled fixed or live biological samples. ISL predicts a range of labels, such as those for nuclei, cell type (e.g., neural), and cell state (e.g., cell death). Because prediction happens in silico, the method is consistent, is not limited by spectral overlap, and does not disturb the experiment. ISL generates biological measurements that would otherwise be problematic or impossible to acquire.


Assuntos
Corantes Fluorescentes/química , Processamento de Imagem Assistida por Computador/métodos , Microscopia de Fluorescência/métodos , Neurônios Motores/citologia , Algoritmos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Córtex Cerebral/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Aprendizado de Máquina , Redes Neurais de Computação , Neurociências , Ratos , Software , Células-Tronco/citologia
2.
Mol Cell ; 70(2): 211-227.e8, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29656925

RESUMO

Flux through kinase and ubiquitin-driven signaling systems depends on the modification kinetics, stoichiometry, primary site specificity, and target abundance within the pathway, yet we rarely understand these parameters and their spatial organization within cells. Here we develop temporal digital snapshots of ubiquitin signaling on the mitochondrial outer membrane in embryonic stem cell-derived neurons, and we model HeLa cell systems upon activation of the PINK1 kinase and PARKIN ubiquitin ligase by proteomic counting of ubiquitylation and phosphorylation events. We define the kinetics and site specificity of PARKIN-dependent target ubiquitylation, and we demonstrate the power of this approach to quantify pathway modulators and to mechanistically define the role of PARKIN UBL phosphorylation in pathway activation in induced neurons. Finally, through modulation of pS65-Ub on mitochondria, we demonstrate that Ub hyper-phosphorylation is inhibitory to mitophagy receptor recruitment, indicating that pS65-Ub stoichiometry in vivo is optimized to coordinate PARKIN recruitment via pS65-Ub and mitophagy receptors via unphosphorylated chains.


Assuntos
Células-Tronco Embrionárias Humanas/enzimologia , Membranas Mitocondriais/enzimologia , Células-Tronco Neurais/enzimologia , Neurogênese , Neurônios/enzimologia , Proteômica/métodos , Ubiquitina-Proteína Ligases/metabolismo , Ativação Enzimática , Células HeLa , Humanos , Cinética , Mitofagia , Fenótipo , Fosforilação , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Canal de Ânion 1 Dependente de Voltagem/genética , Canal de Ânion 1 Dependente de Voltagem/metabolismo
3.
Cell ; 132(4): 549-52, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18295572

RESUMO

Much of the attention focused on stem cells relates to their use in cell replacement therapy; however, stem cells may also transform the way in which therapeutics are discovered and validated.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Células-Tronco Embrionárias/citologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Camundongos
4.
BMC Neurol ; 21(1): 201, 2021 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-34006233

RESUMO

BACKGROUND: Characterization of prediagnostic Parkinson's Disease (PD) and early prediction of subsequent development are critical for preventive interventions, risk stratification and understanding of disease pathology. This study aims to characterize the role of the prediagnostic period in PD and, using selected features from this period as novel interception points, construct a prediction model to accelerate the diagnosis in a real-world setting. METHODS: We constructed two sets of machine learning models: a retrospective approach highlighting exposures up to 5 years prior to PD diagnosis, and an alternative model that prospectively predicted future PD diagnosis from all individuals at their first diagnosis of a gait or tremor disorder, these being features that appeared to represent the initiation of a differential diagnostic window. RESULTS: We found many novel features captured by the retrospective models; however, the high accuracy was primarily driven from surrogate diagnoses for PD, such as gait and tremor disorders, suggesting the presence of a distinctive differential diagnostic period when the clinician already suspected PD. The model utilizing a gait/tremor diagnosis as the interception point, achieved a validation AUC of 0.874 with potential time compression to a future PD diagnosis of more than 300 days. Comparisons of predictive diagnoses between the prospective and prediagnostic cohorts suggest the presence of distinctive trajectories of PD progression based on comorbidity profiles. CONCLUSIONS: Overall, our machine learning approach allows for both guiding clinical decisions such as the initiation of neuroprotective interventions and importantly, the possibility of earlier diagnosis for clinical trials for disease modifying therapies.


Assuntos
Doença de Parkinson/diagnóstico , Marcha/fisiologia , Análise da Marcha , Humanos , Aprendizado de Máquina , Estudos Retrospectivos , Medição de Risco , Tremor
5.
Proc Natl Acad Sci U S A ; 115(36): E8368-E8377, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30120125

RESUMO

Variations in a multitude of material microenvironmental properties have been observed across tissues in vivo, and these have profound effects on cell phenotype. Phenomenological experiments have suggested that certain of these features of the physical microenvironment, such as stiffness, could sensitize cells to other features; meanwhile, mechanistic studies have detailed a number of biophysical mechanisms for this sensing. However, the broad molecular consequences of these potentially complex and nonlinear interactions bridging from biophysical sensing to phenotype have not been systematically characterized, limiting the overall understanding and rational deployment of these biophysical cues. Here, we explore these interactions by employing a 3D cell culture system that allows for the independent control of culture substrate stiffness, stress relaxation, and adhesion ligand density to systematically explore the transcriptional programs affected by distinct combinations of biophysical parameters using RNA-seq. In mouse mesenchymal stem cells and human cortical neuron progenitors, we find dramatic coupling among these substrate properties, and that the relative contribution of each property to changes in gene expression varies with cell type. Motivated by the bioinformatic analysis, the stiffness of hydrogels encapsulating mouse mesenchymal stem cells was found to regulate the secretion of a wide range of cytokines, and to accordingly influence hematopoietic stem cell differentiation in a Transwell coculture model. These results give insights into how biophysical features are integrated by cells across distinct tissues and offer strategies to synthetic biologists and bioengineers for designing responses to a cell's biophysical environment.


Assuntos
Alginatos , Técnicas de Cultura de Células/métodos , Células-Tronco Hematopoéticas/metabolismo , Hidrogéis , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco , Transcrição Gênica/efeitos dos fármacos , Alginatos/química , Alginatos/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Ácido Glucurônico/química , Ácido Glucurônico/farmacologia , Células-Tronco Hematopoéticas/citologia , Ácidos Hexurônicos/química , Ácidos Hexurônicos/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Camundongos
6.
BMC Biol ; 18(1): 127, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32938453

RESUMO

BACKGROUND: Understanding the genetic modifiers of neurodegenerative diseases can provide insight into the mechanisms underlying these disorders. Here, we examine the relationship between the motor neuron disease spinal muscular atrophy (SMA), which is caused by reduced levels of the survival of motor neuron (SMN) protein, and the actin-bundling protein Plastin 3 (PLS3). Increased PLS3 levels suppress symptoms in a subset of SMA patients and ameliorate defects in SMA disease models, but the functional connection between PLS3 and SMN is poorly understood. RESULTS: We provide immunohistochemical and biochemical evidence for large protein complexes localized in vertebrate motor neuron processes that contain PLS3, SMN, and members of the hnRNP F/H family of proteins. Using a Caenorhabditis elegans (C. elegans) SMA model, we determine that overexpression of PLS3 or loss of the C. elegans hnRNP F/H ortholog SYM-2 enhances endocytic function and ameliorates neuromuscular defects caused by decreased SMN-1 levels. Furthermore, either increasing PLS3 or decreasing SYM-2 levels suppresses defects in a C. elegans ALS model. CONCLUSIONS: We propose that hnRNP F/H act in the same protein complex as PLS3 and SMN and that the function of this complex is critical for endocytic pathways, suggesting that hnRNP F/H proteins could be potential targets for therapy development.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Glicoproteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Atrofia Muscular Espinal/genética , Proteínas de Ligação a RNA/genética , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Animais , Animais Geneticamente Modificados/fisiologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Modelos Animais de Doenças , Endocitose/genética , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo
7.
Circ Res ; 118(7): 1125-41; discussion 1142, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27034275

RESUMO

Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor ß superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Fatores de Diferenciação de Crescimento/fisiologia , Miostatina/fisiologia , Adulto , Envelhecimento/fisiologia , Sequência de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/química , Proteínas Morfogenéticas Ósseas/deficiência , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Dimerização , Feminino , Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/metabolismo , Fatores de Diferenciação de Crescimento/química , Fatores de Diferenciação de Crescimento/deficiência , Fatores de Diferenciação de Crescimento/uso terapêutico , Coração/fisiologia , Cardiopatias/metabolismo , Humanos , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Músculos/fisiologia , Miocárdio/metabolismo , Miostatina/química , Miostatina/deficiência , Especificidade de Órgãos , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Relação Estrutura-Atividade
8.
Nat Chem Biol ; 10(8): 632-639, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24952596

RESUMO

The reprogramming of somatic cells to pluripotency using defined transcription factors holds great promise for biomedicine. However, human reprogramming remains inefficient and relies either on the use of the potentially dangerous oncogenes KLF4 and CMYC or the genetic inhibition of the tumor suppressor gene p53. We hypothesized that inhibition of signal transduction pathways that promote differentiation of the target somatic cells during development might relieve the requirement for non-core pluripotency factors during induced pluripotent stem cell (iPSC) reprogramming. Here, we show that inhibition of Notch greatly improves the efficiency of iPSC generation from mouse and human keratinocytes by suppressing p21 in a p53-independent manner and thereby enriching for undifferentiated cells capable of long-term self-renewal. Pharmacological inhibition of Notch enabled routine production of human iPSCs without KLF4 and CMYC while leaving p53 activity intact. Thus, restricting the development of somatic cells by altering intercellular communication enables the production of safer human iPSCs.


Assuntos
Oncogenes/fisiologia , Células-Tronco Pluripotentes/fisiologia , Receptores Notch/antagonistas & inibidores , Animais , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dipeptídeos/farmacologia , Genes myc , Genes p53 , Histona-Lisina N-Metiltransferase , Humanos , Queratinócitos/efeitos dos fármacos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(26): E2371-80, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23757500

RESUMO

The clinical severity of the neurodegenerative disorder spinal muscular atrophy (SMA) is dependent on the levels of functional Survival Motor Neuron (SMN) protein. Consequently, current strategies for developing treatments for SMA generally focus on augmenting SMN levels. To identify additional potential therapeutic avenues and achieve a greater understanding of SMN, we applied in vivo, in vitro, and in silico approaches to identify genetic and biochemical interactors of the Drosophila SMN homolog. We identified more than 300 candidate genes that alter an Smn-dependent phenotype in vivo. Integrating the results from our genetic screens, large-scale protein interaction studies, and bioinformatic analysis, we define a unique interactome for SMN that provides a knowledge base for a better understanding of SMA.


Assuntos
Proteínas de Drosophila/genética , Genes de Insetos , Proteínas de Ligação a RNA/genética , Animais , Animais Geneticamente Modificados , Redes Reguladoras de Genes , Humanos , Bases de Conhecimento , Junção Neuromuscular/genética , Fenótipo , Interferência de RNA , Especificidade da Espécie , Atrofias Musculares Espinais da Infância/genética
10.
Proc Natl Acad Sci U S A ; 109(10): 3915-20, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22345561

RESUMO

Diabetes is a pathological condition characterized by relative insulin deficiency, persistent hyperglycemia, and, consequently, diffuse micro- and macrovascular disease. One therapeutic strategy is to amplify insulin-secretion capacity by increasing the number of the insulin-producing ß cells without triggering a generalized proliferative response. Here, we present the development of a small-molecule screening platform for the identification of molecules that increase ß-cell replication. Using this platform, we identify a class of compounds [adenosine kinase inhibitors (ADK-Is)] that promote replication of primary ß cells in three species (mouse, rat, and pig). Furthermore, the replication effect of ADK-Is is cell type-selective: treatment of islet cell cultures with ADK-Is increases replication of ß cells but not that of α cells, PP cells, or fibroblasts. Short-term in vivo treatment with an ADK-I also increases ß-cell replication but not exocrine cell or hepatocyte replication. Therefore, we propose ADK inhibition as a strategy for the treatment of diabetes.


Assuntos
Adenosina Quinase/farmacologia , Regulação da Expressão Gênica , Células Secretoras de Insulina/citologia , Animais , Feminino , Fibroblastos/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/metabolismo , Hepatócitos/citologia , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Receptores de Glucagon/metabolismo , Suínos , Serina-Treonina Quinases TOR/metabolismo
11.
Nature ; 455(7211): 406-10, 2008 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-18754008

RESUMO

Ligand-dependent activation of the hedgehog (Hh) signalling pathway has been associated with tumorigenesis in a number of human tissues. Here we show that, although previous reports have described a cell-autonomous role for Hh signalling in these tumours, Hh ligands fail to activate signalling in tumour epithelial cells. In contrast, our data support ligand-dependent activation of the Hh pathway in the stromal microenvironment. Specific inhibition of Hh signalling using small molecule inhibitors, a neutralizing anti-Hh antibody or genetic deletion of smoothened (Smo) in the mouse stroma results in growth inhibition in xenograft tumour models. Taken together, these studies demonstrate a paracrine requirement for Hh ligand signalling in the tumorigenesis of Hh-expressing cancers and have important implications for the development of Hh pathway antagonists in cancer.


Assuntos
Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Comunicação Parácrina/fisiologia , Células Estromais/metabolismo , Animais , Linhagem Celular , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened
12.
J Clin Invest ; 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722695

RESUMO

Spinal Muscular Atrophy (SMA) is typically characterized as a motor neuron disease, but extra-neuronal phenotypes are present in almost every organ in severely affected patients and animal models. Extra-neuronal phenotypes were previously underappreciated as patients with severe SMA phenotypes usually died in infancy; however, with current treatments for motor neurons increasing patient lifespan, impaired function of peripheral organs may develop into significant future comorbidities and lead to new treatment-modified phenotypes. Fatty liver is seen in SMA animal models , but generalizability to patients and whether this is due to hepatocyte-intrinsic Survival Motor Neuron (SMN) protein deficiency and/or subsequent to skeletal muscle denervation is unknown. If liver pathology in SMA is SMN-dependent and hepatocyte-intrinsic, this suggests SMN repleting therapies must target extra-neuronal tissues and motor neurons for optimal patient outcome. Here we showed that fatty liver is present in SMA and that SMA patient-specific iHeps were susceptible to steatosis. Using proteomics, functional studies and CRISPR/Cas9 gene editing, we confirmed that fatty liver in SMA is a primary SMN-dependent hepatocyte-intrinsic liver defect associated with mitochondrial and other hepatic metabolism implications. These pathologies require monitoring and indicate need for systematic clinical surveillance and additional and/or combinatorial therapies to ensure continued SMA patient health.

13.
Dev Biol ; 368(2): 323-34, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22705478

RESUMO

Mutations in the Survival of Motor Neuron (SMN) gene underlie the development of spinal muscular atrophy (SMA), which currently represents the leading genetic cause of mortality in infants and toddlers. SMA is characterized by degeneration of spinal cord motor neurons and muscle atrophy. Although SMA is often considered to be a motor neuron disease, accumulating evidence suggests that muscle cells themselves may be affected by low levels of SMN. Here, we examine satellite cells, tissue-resident stem cells that play an essential role in the growth and repair of skeletal muscle, isolated from a severe SMA mouse model (Smn(-/-); SMN2(+/+)). We found similar numbers of satellite cells in the muscles of SMA and wild-type (Smn(+/+); SMN2(+/+)) mice at postnatal day 2 (P2), and, when isolated from skeletal muscle using cell surface marker expression, these cells showed comparable survival and proliferative potential. However, SMA satellite cells differentiate abnormally, revealed by the premature expression of muscle differentiation markers, and, especially, by a reduced efficiency in forming myotubes. These phenotypes suggest a critical role of SMN protein in the intrinsic regulation of muscle differentiation and suggest that abnormal muscle development contributes to the manifestation of SMA symptoms.


Assuntos
Atrofia Muscular Espinal/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular Espinal/genética , Fator de Transcrição PAX7/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo , Sindecanas/metabolismo , Fatores de Tempo
14.
Nat Chem Biol ; 7(8): 544-52, 2011 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-21685895

RESUMO

The motor neuron disease spinal muscular atrophy (SMA) results from mutations that lead to low levels of the ubiquitously expressed protein survival of motor neuron (SMN). An ever-increasing collection of data suggests that therapeutics that elevate SMN may be effective in treating SMA. We executed an image-based screen of annotated chemical libraries and discovered several classes of compounds that were able to increase cellular SMN. Among the most important was the RTK-PI3K-AKT-GSK-3 signaling cascade. Chemical inhibitors of glycogen synthase kinase 3 (GSK-3) and short hairpin RNAs (shRNAs) directed against this target elevated SMN levels primarily by stabilizing the protein. It was particularly notable that GSK-3 chemical inhibitors were also effective in motor neurons, not only in elevating SMN levels, but also in blocking the death that was produced when SMN was acutely reduced by an SMN-specific shRNA. Thus, we have established a screen capable of detecting drug-like compounds that correct the main phenotypic change underlying SMA.


Assuntos
Descoberta de Drogas/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Atrofia Muscular Espinal/tratamento farmacológico , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Adulto , Animais , Benzazepinas/farmacologia , Células Cultivadas , Pré-Escolar , Células-Tronco Embrionárias , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/fisiologia , Inativação Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Indóis/farmacologia , Camundongos , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/metabolismo , Mutação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Transcrição STAT1 , Bibliotecas de Moléculas Pequenas , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo
15.
Proc Natl Acad Sci U S A ; 107(29): 13111-6, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20616011

RESUMO

Defective RNA metabolism is an emerging mechanism involved in ALS pathogenesis and possibly in other neurodegenerative disorders. Here, we show that microRNA (miRNA) activity is essential for long-term survival of postmitotic spinal motor neurons (SMNs) in vivo. Thus, mice that do not process miRNA in SMNs exhibit hallmarks of spinal muscular atrophy (SMA), including sclerosis of the spinal cord ventral horns, aberrant end plate architecture, and myofiber atrophy with signs of denervation. Furthermore, a neurofilament heavy subunit previously implicated in motor neuron degeneration is specifically up-regulated in miRNA-deficient SMNs. We demonstrate that the heavy neurofilament subunit is a target of miR-9, a miRNA that is specifically down-regulated in a genetic model of SMA. These data provide evidence for miRNA function in SMN diseases and emphasize the potential role of miR-9-based regulatory mechanisms in adult neurons and neurodegenerative states.


Assuntos
MicroRNAs/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatologia , Animais , Axônios/metabolismo , Axônios/patologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Camundongos , Camundongos Mutantes , MicroRNAs/genética , Atividade Motora/fisiologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Denervação Muscular , Proteínas de Neurofilamentos/metabolismo , Subunidades Proteicas/metabolismo , Ribonuclease III/metabolismo , Análise de Sobrevida
16.
Bio Protoc ; 13(9): e4666, 2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37188109

RESUMO

Management of neuropathic pain is notoriously difficult; current analgesics, including anti-inflammatory- and opioid-based medications, are generally ineffective and can pose serious side effects. There is a need to uncover non-addictive and safe analgesics to combat neuropathic pain. Here, we describe the setup of a phenotypic screen whereby the expression of an algesic gene,Gch1, is targeted. GCH1 is the rate-limiting enzyme in the de novo synthesis of tetrahydrobiopterin (BH4), a metabolite linked to neuropathic pain in both animal models and in human chronic pain sufferers.Gch1is induced in sensory neurons after nerve injury and its upregulation is responsible for increased BH4 levels. GCH1 protein has proven to be a difficult enzyme to pharmacologically target with small molecule inhibition. Thus, by establishing a platform to monitor and target inducedGch1 expression in individual injured dorsal root ganglion (DRG) neurons in vitro, we can screen for compounds that regulate its expression levels. This approach also allows us to gain valuable biological insights into the pathways and signals regulating GCH1 and BH4 levels upon nerve injury. This protocol is compatible with any transgenic reporter system in which the expression of an algesic gene (or multiple genes) can be monitored fluorescently. Such an approach can be scaled up for high-throughput compound screening and is amenable to transgenic mice as well as human stem cell-derived sensory neurons. Graphical overview.

17.
Front Cell Neurosci ; 17: 1327361, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38314348

RESUMO

Amyotrophic lateral sclerosis (ALS) is a degenerative motor neuron (MN) disease with severely limited treatment options. Identification of effective treatments has been limited in part by the lack of predictive animal models for complex human disorders. Here, we utilized pharmacologic ER stressors to exacerbate underlying sensitivities conferred by ALS patient genetics in induced pluripotent stem cell (iPSC)-derived motor neurons (MNs). In doing so, we found that thapsigargin and tunicamycin exposure recapitulated ALS-associated degeneration, and that we could rescue this degeneration via MAP4K4 inhibition (MAP4K4i). We subsequently identified mechanisms underlying MAP4K4i-mediated protection by performing phosphoproteomics on iPSC-derived MNs treated with ER stressors ±MAP4K4i. Through these analyses, we found JNK, PKC, and BRAF to be differentially modulated in MAP4K4i-protected MNs, and that inhibitors to these proteins could also rescue MN toxicity. Collectively, this study highlights the value of utilizing ER stressors in ALS patient MNs to identify novel druggable targets.

18.
J Cardiovasc Aging ; 3(4)2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38235060

RESUMO

Since the exogenous administration of GDF11, a TGF-ß superfamily member, was reported to have beneficial effects in some models of human disease, there have been many research studies in GDF11 biology. However, many studies have now confirmed that exogenous administration of GDF11 can improve physiology in disease models, including cardiac fibrosis, experimental stroke, and disordered metabolism. GDF11 is similar to GDF8 (also called Myostatin), differing only by 11 amino acids in their mature signaling domains. These two proteins are now known to be biochemically different both in vitro and in vivo. GDF11 is much more potent than GDF8 and induces more strongly SMAD2 phosphorylation in the myocardium compared to GDF8. GDF8 and GDF11 prodomain are only 52% identical and are cleaved by different Tolloid proteases to liberate the mature signaling domain from inhibition of the prodomain. Here, we review the state of GDF11 biology, highlighting both resolved and remaining controversies.

19.
Elife ; 122023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37083703

RESUMO

Resolving fundamental molecular and functional processes underlying human synaptic development is crucial for understanding normal brain function as well as dysfunction in disease. Based upon increasing evidence of species-divergent features of brain cell types, coupled with emerging studies of complex human disease genetics, we developed the first automated and quantitative high-content synaptic phenotyping platform using human neurons and astrocytes. To establish the robustness of our platform, we screened the effects of 376 small molecules on presynaptic density, neurite outgrowth, and cell viability, validating six small molecules that specifically enhanced human presynaptic density in vitro. Astrocytes were essential for mediating the effects of all six small molecules, underscoring the relevance of non-cell-autonomous factors in synapse assembly and their importance in synaptic screening applications. Bromodomain and extraterminal (BET) inhibitors emerged as the most prominent hit class and global transcriptional analyses using multiple BET inhibitors confirmed upregulation of synaptic gene expression. Through these analyses, we demonstrate the robustness of our automated screening platform for identifying potent synaptic modulators, which can be further leveraged for scaled analyses of human synaptic mechanisms and drug discovery efforts.


Assuntos
Neurogênese , Neurônios , Humanos , Neurogênese/fisiologia , Neurônios/fisiologia , Sinapses/fisiologia , Crescimento Neuronal , Astrócitos
20.
iScience ; 26(7): 106995, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37534135

RESUMO

Emerging evidence of species divergent features of astrocytes coupled with the relative inaccessibility of human brain tissue underscore the utility of human pluripotent stem cell (hPSC) technologies for the generation and study of human astrocytes. However, existing approaches for hPSC-astrocyte generation are typically lengthy or require intermediate purification steps. Here, we establish a rapid and highly scalable method for generating functional human induced astrocytes (hiAs). These hiAs express canonical astrocyte markers, respond to pro-inflammatory stimuli, exhibit ATP-induced calcium transients and support neuronal network development. Moreover, single-cell transcriptomic analyses reveal the generation of highly reproducible cell populations across individual donors, mostly resembling human fetal astrocytes. Finally, hiAs generated from a trisomy 21 disease model identify expected alterations in cell-cell adhesion and synaptic signaling, supporting their utility for disease modeling applications. Thus, hiAs provide a valuable and practical resource for the study of basic human astrocyte function and dysfunction in disease.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA