Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(19): E1752-60, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23613578

RESUMEN

A growing body of evidence in humans implicates chronic activation of the innate immune response in the brain as a major cause of neuropathology in various neurodegenerative conditions, although the mechanisms remain unclear. In an unbiased genetic screen for mutants exhibiting neurodegeneration in Drosophila, we have recovered a mutation of dnr1 (defense repressor 1), a negative regulator of the Imd (immune deficiency) innate immune-response pathway. dnr1 mutants exhibit shortened lifespan and progressive, age-dependent neuropathology associated with activation of the Imd pathway and elevated expression of AMP (antimicrobial peptide) genes. To test the hypothesis that overactivation of innate immune-response pathways in the brain is responsible for neurodegeneration, we demonstrated that direct bacterial infection in the brain of wild-type flies also triggers neurodegeneration. In both cases, neurodegeneration is dependent on the NF-κB transcription factor, Relish. Moreover, we found that neural overexpression of individual AMP genes is sufficient to cause neurodegeneration. These results provide a mechanistic link between innate immune responses and neurodegeneration and may have important implications for the role of neuroinflammation in human neurodegenerative diseases as well.


Asunto(s)
Encéfalo/patología , Proteínas de Drosophila/genética , Drosophila melanogaster/inmunología , Inmunidad Innata , Mutación , Proteínas Represoras/genética , Envejecimiento , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Apoptosis , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Encéfalo/inmunología , Encéfalo/microbiología , Muerte Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/microbiología , Genotipo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Fenotipo , Factores de Transcripción/metabolismo
2.
Proc Natl Acad Sci U S A ; 110(44): E4152-9, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24127584

RESUMEN

Traumatic brain injury (TBI) is a substantial health issue worldwide, yet the mechanisms responsible for its complex spectrum of pathologies remains largely unknown. To investigate the mechanisms underlying TBI pathologies, we developed a model of TBI in Drosophila melanogaster. The model allows us to take advantage of the wealth of experimental tools available in flies. Closed head TBI was inflicted with a mechanical device that subjects flies to rapid acceleration and deceleration. Similar to humans with TBI, flies with TBI exhibited temporary incapacitation, ataxia, activation of the innate immune response, neurodegeneration, and death. Our data indicate that TBI results in death shortly after a primary injury only if the injury exceeds a certain threshold and that age and genetic background, but not sex, substantially affect this threshold. Furthermore, this threshold also appears to be dependent on the same cellular and molecular mechanisms that control normal longevity. This study demonstrates the potential of flies for providing key insights into human TBI that may ultimately provide unique opportunities for therapeutic intervention.


Asunto(s)
Aceleración/efectos adversos , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Drosophila melanogaster , Inmunidad Innata/fisiología , Longevidad/fisiología , Factores de Edad , Análisis de Varianza , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/inmunología , Femenino , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores Sexuales
3.
Proc Natl Acad Sci U S A ; 109(11): E656-64, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22355133

RESUMEN

To investigate the mechanistic basis for central nervous system (CNS) neurodegeneration in the disease ataxia-telangiectasia (A-T), we analyzed flies mutant for the causative gene A-T mutated (ATM). ATM encodes a protein kinase that functions to monitor the genomic integrity of cells and control cell cycle, DNA repair, and apoptosis programs. Mutation of the C-terminal amino acid in Drosophila ATM inhibited the kinase activity and caused neuron and glial cell death in the adult brain and a reduction in mobility and longevity. These data indicate that reduced ATM kinase activity is sufficient to cause neurodegeneration in A-T. ATM kinase mutant flies also had elevated expression of innate immune response genes in glial cells. ATM knockdown in glial cells, but not neurons, was sufficient to cause neuron and glial cell death, a reduction in mobility and longevity, and elevated expression of innate immune response genes in glial cells, indicating that a non-cell-autonomous mechanism contributes to neurodegeneration in A-T. Taken together, these data suggest that early-onset CNS neurodegeneration in A-T is similar to late-onset CNS neurodegeneration in diseases such as Alzheimer's in which uncontrolled inflammatory response mediated by glial cells drives neurodegeneration.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Drosophila/antagonistas & inhibidores , Drosophila melanogaster/enzimología , Drosophila melanogaster/inmunología , Inmunidad Innata/inmunología , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/inmunología , Neuroglía/enzimología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Muerte Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Técnicas de Silenciamiento del Gen , Inmunidad Innata/genética , Longevidad , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuroglía/inmunología , Neuroglía/patología , Neuronas/enzimología , Neuronas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Temperatura , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba/genética
4.
Nat Biotechnol ; 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974010

RESUMEN

Central norepinephrine (NE) neurons, located mainly in the locus coeruleus (LC), are implicated in diverse psychiatric and neurodegenerative diseases and are an emerging target for drug discovery. To facilitate their study, we developed a method to generate 40-60% human LC-NE neurons from human pluripotent stem cells. The approach depends on our identification of ACTIVIN A in regulating LC-NE transcription factors in dorsal rhombomere 1 (r1) progenitors. In vitro generated human LC-NE neurons display extensive axonal arborization; release and uptake NE; and exhibit pacemaker activity, calcium oscillation and chemoreceptor activity in response to CO2. Single-nucleus RNA sequencing (snRNA-seq) analysis at multiple timepoints confirmed NE cell identity and revealed the differentiation trajectory from hindbrain progenitors to NE neurons via an ASCL1-expressing precursor stage. LC-NE neurons engineered with an NE sensor reliably reported extracellular levels of NE. The availability of functional human LC-NE neurons enables investigation of their roles in psychiatric and neurodegenerative diseases and provides a tool for therapeutics development.

5.
Aging Cell ; 21(1): e13541, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34953016

RESUMEN

Modeling age-related neurodegenerative disorders with human stem cells are difficult due to the embryonic nature of stem cell-derived neurons. We developed a chemical cocktail to induce senescence of iPSC-derived neurons to address this challenge. We first screened small molecules that induce embryonic fibroblasts to exhibit features characteristic of aged fibroblasts. We then optimized a cocktail of small molecules that induced senescence in fibroblasts and cortical neurons without causing DNA damage. The utility of the "senescence cocktail" was validated in motor neurons derived from ALS patient iPSCs which exhibited protein aggregation and axonal degeneration substantially earlier than those without cocktail treatment. Our "senescence cocktail" will likely enhance the manifestation of disease-related phenotypes in neurons derived from iPSCs, enabling the generation of reliable drug discovery platforms.


Asunto(s)
Neuronas Motoras/metabolismo , Enfermedades Neurodegenerativas/genética , Diferenciación Celular , Humanos , Fenotipo
6.
Stem Cell Reports ; 16(6): 1446-1457, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33861989

RESUMEN

Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) and their differentiation into neural lineages is a revolutionary experimental system for studying neurological disorders, including intellectual and developmental disabilities (IDDs). However, issues related to variability and reproducibility have hindered translating preclinical findings into drug discovery. Here, we identify areas for improvement by conducting a comprehensive review of 58 research articles that utilized iPSC-derived neural cells to investigate genetically defined IDDs. Based upon these findings, we propose recommendations for best practices that can be adopted by research scientists as well as journal editors.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Variación Genética , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual/etiología , Humanos , Modelos Biológicos , Neuronas , Reproducibilidad de los Resultados
7.
Endocrinology ; 162(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34125902

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus play a key role in the regulation of reproductive function. In this study, we sought an efficient method for generating GnRH neurons from human embryonic and induced pluripotent stem cells (hESC and hiPSC, respectively). First, we found that exposure of primitive neuroepithelial cells, rather than neuroprogenitor cells, to fibroblast growth factor 8 (FGF8), was more effective in generating GnRH neurons. Second, addition of kisspeptin to FGF8 further increased the efficiency rates of GnRH neurogeneration. Third, we generated a fluorescent marker mCherry labeled human embryonic GnRH cell line (mCh-hESC) using a CRISPR-Cas9 targeting approach. Fourth, we examined physiological characteristics of GnRH (mCh-hESC) neurons: similar to GnRH neurons in vivo, they released the GnRH peptide in a pulsatile manner at ~60 min intervals; GnRH release increased in response to high potassium, kisspeptin, estradiol, and neurokinin B challenges; and injection of depolarizing current induced action potentials. Finally, we characterized developmental changes in transcriptomes of GnRH neurons using hESC, hiPSC, and mCh-hESC. The developmental pattern of transcriptomes was remarkably similar among the 3 cell lines. Collectively, human stem cell-derived GnRH neurons will be an important tool for establishing disease models to understand diseases, such as idiopathic hypothalamic hypogonadism, and testing contraceptive drugs.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Células Madre Embrionarias Humanas/fisiología , Neurogénesis/genética , Neuronas/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Factor 8 de Crecimiento de Fibroblastos/farmacología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/fisiología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Transcriptoma/efectos de los fármacos
9.
Stem Cell Res ; 34: 101365, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30605841

RESUMEN

Seven human induced pluripotent stem cell (iPSC) lines were generated from fibroblasts from three neonatal individuals using non-integrative reprogramming. Most control iPSCs are derived from adults, so these iPSCs meet the need for control iPSCs from young individuals. Donors were from different ethnicities and these lines provide unique genetic profiles. All iPSCs have normal karyotypes, express stem cell markers, and exhibit pluripotency, as assessed by capacity to differentiate into three germ layers. These lines are valuable to study human development, as age-matched controls for disorder-specific iPSCs, and as platforms for gene editing to control for age and ethnicity.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Etnicidad , Animales , Línea Celular , Femenino , Humanos , Recién Nacido , Masculino , Ratones
10.
Nat Commun ; 10(1): 53, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30604771

RESUMEN

CRISPR/Cas9 guided gene-editing is a potential therapeutic tool, however application to neurodegenerative disease models has been limited. Moreover, conventional mutation correction by gene-editing would only be relevant for the small fraction of neurodegenerative cases that are inherited. Here we introduce a CRISPR/Cas9-based strategy in cell and animal models to edit endogenous amyloid precursor protein (APP) at the extreme C-terminus and reciprocally manipulate the amyloid pathway, attenuating APP-ß-cleavage and Aß production, while up-regulating neuroprotective APP-α-cleavage. APP N-terminus and compensatory APP-homologues remain intact, with no apparent effects on neurophysiology in vitro. Robust APP-editing is seen in human iPSC-derived neurons and mouse brains with no detectable off-target effects. Our strategy likely works by limiting APP and BACE-1 approximation, and we also delineate mechanistic events that abrogates APP/BACE-1 convergence in this setting. Our work offers conceptual proof for a selective APP silencing strategy.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Edición Génica/métodos , Terapia Genética/métodos , Enfermedades Neurodegenerativas/terapia , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/citología , Encéfalo/patología , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas , Técnicas Estereotáxicas , Transfección , Resultado del Tratamiento
11.
Stem Cell Reports ; 9(6): 1885-1897, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29153989

RESUMEN

Alzheimer's disease (AD) is a common neurodegenerative disorder and the leading cause of cognitive impairment. Due to insufficient understanding of the disease mechanisms, there are no efficient therapies for AD. Most studies have focused on neuronal cells, but astrocytes have also been suggested to contribute to AD pathology. We describe here the generation of functional astrocytes from induced pluripotent stem cells (iPSCs) derived from AD patients with PSEN1 ΔE9 mutation, as well as healthy and gene-corrected isogenic controls. AD astrocytes manifest hallmarks of disease pathology, including increased ß-amyloid production, altered cytokine release, and dysregulated Ca2+ homeostasis. Furthermore, due to altered metabolism, AD astrocytes show increased oxidative stress and reduced lactate secretion, as well as compromised neuronal supportive function, as evidenced by altering Ca2+ transients in healthy neurons. Our results reveal an important role for astrocytes in AD pathology and highlight the strength of iPSC-derived models for brain diseases.


Asunto(s)
Enfermedad de Alzheimer/genética , Mitocondrias/metabolismo , Neuronas/patología , Presenilina-1/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/biosíntesis , Péptidos beta-Amiloides/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Calcio/metabolismo , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Ácido Láctico/metabolismo , Mitocondrias/patología , Neuronas/metabolismo , Estrés Oxidativo/genética
12.
Cell Stem Cell ; 17(2): 233-44, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26145478

RESUMEN

Precise temporal control of gene expression or deletion is critical for elucidating gene function in biological systems. However, the establishment of human pluripotent stem cell (hPSC) lines with inducible gene knockout (iKO) remains challenging. We explored building iKO hPSC lines by combining CRISPR/Cas9-mediated genome editing with the Flp/FRT and Cre/LoxP system. We found that "dual-sgRNA targeting" is essential for biallelic knockin of FRT sequences to flank the exon. We further developed a strategy to simultaneously insert an activity-controllable recombinase-expressing cassette and remove the drug-resistance gene, thus speeding up the generation of iKO hPSC lines. This two-step strategy was used to establish human embryonic stem cell (hESC) and induced pluripotent stem cell (iPSC) lines with iKO of SOX2, PAX6, OTX2, and AGO2, genes that exhibit diverse structural layout and temporal expression patterns. The availability of iKO hPSC lines will substantially transform the way we examine gene function in human cells.


Asunto(s)
Sistemas CRISPR-Cas/genética , Técnicas de Inactivación de Genes , Ingeniería Genética/métodos , Células Madre/metabolismo , Secuencia de Bases , Diferenciación Celular , Exones/genética , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Marcación de Gen , Homocigoto , Humanos , Datos de Secuencia Molecular , Mutagénesis Insercional/genética , Factores de Transcripción Otx/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Células Madre Pluripotentes/metabolismo , ARN Guía de Kinetoplastida/metabolismo
13.
Genetics ; 194(1): 133-42, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23502677

RESUMEN

Neurodegeneration is a hallmark of the human disease ataxia-telangiectasia (A-T) that is caused by mutation of the A-T mutated (ATM) gene. We have analyzed Drosophila melanogaster ATM mutants to determine the molecular mechanisms underlying neurodegeneration in A-T. Previously, we found that ATM mutants upregulate the expression of innate immune response (IIR) genes and undergo neurodegeneration in the central nervous system. Here, we present evidence that activation of the IIR is a cause of neurodegeneration in ATM mutants. Three lines of evidence indicate that ATM mutations cause neurodegeneration by activating the Nuclear Factor-κB (NF-κB) transcription factor Relish, a key regulator of the Immune deficiency (Imd) IIR signaling pathway. First, the level of upregulation of IIR genes, including Relish target genes, was directly correlated with the level of neurodegeneration in ATM mutants. Second, Relish mutations inhibited upregulation of IIR genes and neurodegeneration in ATM mutants. Third, overexpression of constitutively active Relish in glial cells activated the IIR and caused neurodegeneration. In contrast, we found that Imd and Dif mutations did not affect neurodegeneration in ATM mutants. Imd encodes an activator of Relish in the response to gram-negative bacteria, and Dif encodes an immune responsive NF-κB transcription factor in the Toll signaling pathway. These data indicate that the signal that causes neurodegeneration in ATM mutants activates a specific NF-κB protein and does so through an unknown activator. In summary, these findings suggest that neurodegeneration in human A-T is caused by activation of a specific NF-κB protein in glial cells.


Asunto(s)
Ataxia Telangiectasia/inmunología , Ataxia Telangiectasia/patología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Inmunidad Innata , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Factores de Transcripción/metabolismo , Animales , Ataxia Telangiectasia/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Encéfalo/patología , Proteínas de Ciclo Celular/genética , Muerte Celular , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/genética , Longevidad , Actividad Motora , Mutación/genética , Degeneración Nerviosa/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
14.
Fly (Austin) ; 6(3): 169-72, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22864563

RESUMEN

In this Extra View, we highlight recent Drosophila research that has uncovered a new role for the innate immune response. The research indicates that, in addition to combating infection, the innate immune response promotes neurodegeneration. Our publication (Petersen et al., 2012) reveals a correlative relationship between the innate immune response and neurodegeneration in a model of the human disease Ataxia-telangiectasia (A-T). We also found that glial cells are responsible for the innate immune response in the A-T model, and work by others implicates glial cells in neurodegeneration. Additionally, publications by Chinchore et al. (2012) and Tan et al. (2008) reveal a causative role for the innate immune response in models of human retinal degenerative disorders and Alzheimer disease, respectively. Collectively, these findings suggest that activation of the innate immune response is a shared cause of neurodegeneration in different human diseases.


Asunto(s)
Drosophila/inmunología , Inmunidad Innata , Degeneración Nerviosa/inmunología , Animales , Ataxia Telangiectasia/inmunología , Ataxia Telangiectasia/patología , Modelos Animales de Enfermedad , Humanos , Modelos Inmunológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA