Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1169962, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37384248

RESUMEN

Aggregation of the Tar DNA-binding protein of 43 kDa (TDP-43) is a pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia and likely contributes to disease by loss of nuclear function. Analysis of TDP-43 function in knockout zebrafish identified an endothelial directional migration and hypersprouting phenotype during development prior lethality. In human umbilical vein cells (HUVEC) the loss of TDP-43 leads to hyperbranching. We identified elevated expression of FIBRONECTIN 1 (FN1), the VASCULAR CELL ADHESION MOLECULE 1 (VCAM1), as well as their receptor INTEGRIN α4ß1 (ITGA4B1) in HUVEC cells. Importantly, reducing the levels of ITGA4, FN1, and VCAM1 homologues in the TDP-43 loss-of-function zebrafish rescues the angiogenic defects indicating the conservation of human and zebrafish TDP-43 function during angiogenesis. Our study identifies a novel pathway regulated by TDP-43 important for angiogenesis during development.

2.
Mol Metab ; 74: 101748, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37290673

RESUMEN

OBJECTIVE: Cancer cells convert more glucose into lactate than healthy cells, what contributes to their growth advantage. Pyruvate kinase (PK) is a key rate limiting enzyme in this process, what makes it a promising potential therapeutic target. However, currently it is still unclear what consequences the inhibition of PK has on cellular processes. Here, we systematically investigate the consequences of PK depletion for gene expression, histone modifications and metabolism. METHODS: Epigenetic, transcriptional and metabolic targets were analysed in different cellular and animal models with stable knockdown or knockout of PK. RESULTS: Depleting PK activity reduces the glycolytic flux and causes accumulation of glucose-6-phosphate (G6P). Such metabolic perturbation results in stimulation of the activity of a heterodimeric pair of transcription factors MondoA and MLX but not in a major reprogramming of the global H3K9ac and H3K4me3 histone modification landscape. The MondoA:MLX heterodimer upregulates expression of thioredoxin-interacting protein (TXNIP) - a tumour suppressor with multifaceted anticancer activity. This effect of TXNIP upregulation extends beyond immortalised cancer cell lines and is applicable to multiple cellular and animal models. CONCLUSIONS: Our work shows that actions of often pro-tumorigenic PK and anti-tumorigenic TXNIP are tightly linked via a glycolytic intermediate. We suggest that PK depletion stimulates the activity of MondoA:MLX transcription factor heterodimers and subsequently, increases cellular TXNIP levels. TXNIP-mediated inhibition of thioredoxin (TXN) can reduce the ability of cells to scavenge reactive oxygen species (ROS) leading to the oxidative damage of cellular structures including DNA. These findings highlight an important regulatory axis affecting tumour suppression mechanisms and provide an attractive opportunity for combination cancer therapies targeting glycolytic activity and ROS-generating pathways.


Asunto(s)
Neoplasias , Piruvato Quinasa , Animales , Piruvato Quinasa/genética , Especies Reactivas de Oxígeno , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Tiorredoxinas/química , Tiorredoxinas/metabolismo
3.
Mol Neurodegener ; 12(1): 6, 2017 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-28088213

RESUMEN

BACKGROUND: The most frequent genetic cause of frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) is the expansion of a GGGGCC hexanucleotide repeat in a non-coding region of the chromosome 9 open reading frame 72 (C9orf72) locus. The pathological hallmarks observed in C9orf72 repeat expansion carriers are the formation of RNA foci and deposition of dipeptide repeat (DPR) proteins derived from repeat associated non-ATG (RAN) translation. Currently, it is unclear whether formation of RNA foci, DPR translation products, or partial loss of C9orf72 predominantly drive neurotoxicity in vivo. By using a transgenic approach in zebrafish we address if the most frequently found DPR in human ALS/FTLD brain, the poly-Gly-Ala (poly-GA) protein, is toxic in vivo. METHOD: We generated several transgenic UAS responder lines that express either 80 repeats of GGGGCC alone, or together with a translation initiation ATG codon forcing the translation of GA80-GFP protein upon crossing to a Gal4 driver. The GGGGCC repeat and GA80 were fused to green fluorescent protein (GFP) lacking a start codon to monitor protein translation by GFP fluorescence. RESULTS: Zebrafish transgenic for the GGGGCC repeat lacking an ATG codon showed very mild toxicity in the absence of poly-GA. However, strong toxicity was induced upon ATG initiated expression of poly-GA, which was rescued by injection of an antisense morpholino interfering with start codon dependent poly-GA translation. This morpholino only interferes with GA80-GFP translation without affecting repeat transcription, indicating that the toxicity is derived from GA80-GFP. CONCLUSION: These novel transgenic C9orf72 associated repeat zebrafish models demonstrate poly-GA toxicity in zebrafish. Reduction of poly-GA protein rescues toxicity validating this therapeutic approach to treat C9orf72 repeat expansion carriers. These novel animal models provide a valuable tool for drug discovery to reduce DPR associated toxicity in ALS/FTLD patients with C9orf72 repeat expansions.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Degeneración Lobar Frontotemporal/genética , Sistemas de Lectura Abierta , Péptidos/toxicidad , Esclerosis Amiotrófica Lateral/patología , Animales , Animales Modificados Genéticamente , Western Blotting , Repeticiones de Dinucleótido , Modelos Animales de Enfermedad , Degeneración Lobar Frontotemporal/patología , Inmunohistoquímica , Hibridación Fluorescente in Situ , Reacción en Cadena de la Polimerasa , Polímeros , Pez Cebra
4.
Methods Mol Biol ; 1254: 341-50, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25431076

RESUMEN

The CRISPR /Cas system identified in archaea has been adopted and optimized for genome editing purposes in zebrafish. In vitro transcribed guide RNA and Cas9 mRNA are microinjected into fertilized zebrafish embryos to edit the zebrafish genome. Here, we describe how to design a gRNA, a fast method for in vitro transcription of gRNA from oligonucleotides , microinjection into fertilized zebrafish embryos, and a PCR -based restriction fragment length assay to identify mutations at the gRNA target site.


Asunto(s)
Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas , Edición Génica/métodos , ARN Guía de Kinetoplastida/genética , Pez Cebra/genética , Animales , Genoma , Modelos Animales , Mutación
5.
Development ; 140(24): 4982-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24257628

RESUMEN

Gene modifications in animal models have been greatly facilitated through the application of targeted genome editing tools. The prokaryotic CRISPR/Cas9 type II genome editing system has recently been applied in cell lines and vertebrates. However, we still have very limited information about the efficiency of mutagenesis, germline transmission rates and off-target effects in genomes of model organisms. We now demonstrate that CRISPR/Cas9 mutagenesis in zebrafish is highly efficient, reaching up to 86.0%, and is heritable. The efficiency of the CRISPR/Cas9 system further facilitated the targeted knock-in of a protein tag provided by a donor oligonucleotide with knock-in efficiencies of 3.5-15.6%. Mutation rates at potential off-target sites are only 1.1-2.5%, demonstrating the specificity of the CRISPR/Cas9 system. The ease and efficiency of the CRISPR/Cas9 system with limited off-target effects make it a powerful genome engineering tool for in vivo studies.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas , Endonucleasas/genética , Edición Génica/métodos , Pez Cebra/genética , Animales , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Técnicas de Sustitución del Gen , Genoma , Mutagénesis , Oligonucleótidos Antisentido/genética , Análisis de Secuencia de ADN
6.
Proc Natl Acad Sci U S A ; 110(13): 4986-91, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23457265

RESUMEN

Mutations in the Tar DNA binding protein of 43 kDa (TDP-43; TARDBP) are associated with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43(+) inclusions (FTLD-TDP). To determine the physiological function of TDP-43, we knocked out zebrafish Tardbp and its paralogue Tardbp (TAR DNA binding protein-like), which lacks the glycine-rich domain where ALS- and FTLD-TDP-associated mutations cluster. tardbp mutants show no phenotype, a result of compensation by a unique splice variant of tardbpl that additionally contains a C-terminal elongation highly homologous to the glycine-rich domain of tardbp. Double-homozygous mutants of tardbp and tardbpl show muscle degeneration, strongly reduced blood circulation, mispatterning of vessels, impaired spinal motor neuron axon outgrowth, and early death. In double mutants the muscle-specific actin binding protein Filamin Ca is up-regulated. Strikingly, Filamin C is similarly increased in the frontal cortex of FTLD-TDP patients, suggesting aberrant expression in smooth muscle cells and TDP-43 loss-of-function as one underlying disease mechanism.


Asunto(s)
Axones/metabolismo , Proteínas de Unión al ADN , Neuronas Motoras/metabolismo , Atrofia Muscular/metabolismo , Mutación , Enfermedades Vasculares/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/patología , Proteínas Contráctiles/genética , Proteínas Contráctiles/metabolismo , Filaminas , Humanos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neuronas Motoras/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Estructura Terciaria de Proteína , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología , Proteínas de Pez Cebra/genética
7.
J Neurochem ; 127(4): 471-81, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23406323

RESUMEN

Alzheimer's disease is the most frequent dementia. Pathologically, Alzheimer's disease is characterized by the accumulation of senile plaques composed of amyloid ß-peptide (Aß). Two proteases, ß- and γ-secretase proteolytically generate Aß from its precursor, the ß-amyloid precursor protein (APP). Inhibition of ß-secretase, also referred to as beta-site APP cleaving enzyme (BACE1) or γ-secretase is therefore of prime interest for the development of amyloid-lowering drugs. To assess the in vivo function of zebrafish Bace1 (zBace1), we generated zBace1 knock out fish by zinc finger nuclease-mediated genome editing. bace1 mutants (bace1-/-) are hypomyelinated in the PNS while the CNS is not affected. Moreover, the number of mechanosensory neuromasts is elevated in bace1-/-. Mutations in zebrafish Bace2 (zBace2) revealed a distinct melanocyte migration phenotype, which is not observed in bace1-/-. Double homozygous bace1-/-; bace2-/- fish do not enhance the single mutant phenotypes indicating non-redundant distinct physiological functions. Single homozygous bace1 mutants as well as double homozygous bace1 and bace2 mutants are viable and fertile suggesting that Bace1 is a promising drug target without major side effects. The identification of a specific bace2 -/- associated phenotype further allows improving selective Bace1 inhibitors and to distinguish between Bace 1 and Bace 2 inhibition in vivo. Inhibition of BACE1 protease activity has therapeutic importance for Alzheimer's disease. Analysis of BACE1 and BACE2 knock-out zebrafish revealed that they exhibit distinct phenotypes. bace1 mutants display hypomyelination in the PNS and supernumerary neuromasts while in bace2 mutants the shape and migration of melanocytes is affected. These phenotypes are not further enhanced in the viable double mutants. Our data suggest that blocking BACE1 activity is a safe therapeutic approach.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Melanocitos/fisiología , Proteínas de Pez Cebra/genética , Pez Cebra/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Movimiento Celular , Técnicas de Inactivación de Genes , Larva/genética , Larva/metabolismo , Modelos Animales , Mutación , Fenotipo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/metabolismo
8.
J Neurosci ; 32(46): 16203-12, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-23152604

RESUMEN

Mitochondria provide ATP, maintain calcium homeostasis, and regulate apoptosis. Neurons, due to their size and complex geometry, are particularly dependent on the proper functioning and distribution of mitochondria. Thus disruptions of these organelles and their transport play a central role in a broad range of neurodegenerative diseases. While in vitro studies have greatly expanded our knowledge of mitochondrial dynamics, our understanding in vivo remains limited. To address this shortcoming, we developed tools to study mitochondrial dynamics in vivo in optically accessible zebrafish. We demonstrate here that our newly generated tools, including transgenic "MitoFish," can be used to study the in vivo "life cycle" of mitochondria and allows identifying pharmacological and genetic modulators of mitochondrial dynamics. Furthermore we observed profound mitochondrial transport deficits in real time in a zebrafish tauopathy model. By rescuing this phenotype using MARK2 (microtubule-affinity regulating kinase 2), we provide direct in vivo evidence that this kinase regulates axonal transport in a Tau-dependent manner. Thus, our approach allows detailed studies of the dynamics of mitochondria in their natural environment under normal and disease conditions.


Asunto(s)
Mitocondrias/patología , Enfermedades del Sistema Nervioso/patología , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Antineoplásicos/farmacología , Transporte Biológico/fisiología , Western Blotting , Procesamiento de Imagen Asistido por Computador , Mitocondrias/ultraestructura , Nocodazol/farmacología , Fenotipo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Células Receptoras Sensoriales/fisiología , Células Receptoras Sensoriales/ultraestructura , Proteínas tau/genética
9.
EMBO J ; 29(16): 2841-57, 2010 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-20606625

RESUMEN

Mutations in fused in sarcoma (FUS) are a cause of familial amyotrophic lateral sclerosis (fALS). Patients carrying point mutations in the C-terminus of FUS show neuronal cytoplasmic FUS-positive inclusions, whereas in healthy controls, FUS is predominantly nuclear. Cytoplasmic FUS inclusions have also been identified in a subset of frontotemporal lobar degeneration (FTLD-FUS). We show that a non-classical PY nuclear localization signal (NLS) in the C-terminus of FUS is necessary for nuclear import. The majority of fALS-associated mutations occur within the NLS and impair nuclear import to a degree that correlates with the age of disease onset. This presents the first case of disease-causing mutations within a PY-NLS. Nuclear import of FUS is dependent on Transportin, and interference with this transport pathway leads to cytoplasmic redistribution and recruitment of FUS into stress granules. Moreover, proteins known to be stress granule markers co-deposit with inclusions in fALS and FTLD-FUS patients, implicating stress granule formation in the pathogenesis of these diseases. We propose that two pathological hits, namely nuclear import defects and cellular stress, are involved in the pathogenesis of FUS-opathies.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Carioferinas/metabolismo , Mutación Puntual , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Células Cultivadas , Gránulos Citoplasmáticos/patología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Carioferinas/genética , Datos de Secuencia Molecular , Neuronas/patología , Estructura Terciaria de Proteína , Proteína FUS de Unión a ARN/análisis , Proteína FUS de Unión a ARN/química , Pez Cebra/embriología
10.
Neurobiol Dis ; 39(3): 265-71, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20381619

RESUMEN

Methylene blue is an FDA approved compound with a variety of pharmacologic activities. It inhibits aggregation of several amyloidogenic proteins known to be deposited in neurodegenerative diseases. Recently, it has been proposed that methylene blue shows significant beneficial effects in a phase 2 clinical trial by slowing cognitive decline in Alzheimer's disease patients. To analyze its therapeutic potential, we investigated the effect of methylene blue on neurotoxicity in a zebrafish model for tauopathies. Transgenic expression of the frontotemporal dementia associated Tau-P301L mutation recapitulates a number of the pathological features observed in humans including abnormal phosphorylation and folding of Tau, tangle formation and Tau dependent neuronal loss. Upon incubation of zebrafish larvae with methylene blue, neither abnormal phosphorylation nor neuronal cell loss, reduced neurite outgrowth or a swimming defect were rescued. Methylene blue is biologically active in zebrafish since it reduced aggregation of a huntingtin variant containing a stretch of 102 glutamine residues. However, although huntingtin aggregation was largely prevented by methylene blue, huntingtin-dependent toxicity was unaffected. Our findings are consistent with the hypothesis that toxicity is not necessarily associated with deposition of insoluble amyloid proteins.


Asunto(s)
Muerte Celular/efectos de los fármacos , Azul de Metileno/farmacología , Neuronas/efectos de los fármacos , Péptidos/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Axones/efectos de los fármacos , Western Blotting , Técnica del Anticuerpo Fluorescente , Neuronas/metabolismo , Péptidos/genética , Fosforilación/efectos de los fármacos , Tauopatías/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas tau/genética
11.
J Biol Chem ; 283(3): 1744-1753, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-17984093

RESUMEN

Loss of function mutations in progranulin cause tau-negative frontotemporal lobar degeneration with ubiquitin-positive inclusions. A major protein component of these inclusions is TDP-43, which becomes hyperphosphorylated, ubiquitinated, and cleaved to generate C-terminal fragments, which apparently translocate from nuclei to the cytoplasm. Most progranulin mutations are nonsense mutations resulting in nonsense-mediated mRNA decay and consequently reduced progranulin protein levels. However, some missense mutations are described that occur within the signal sequence and mature progranulin. We now demonstrate that a progranulin mutation located within the signal sequence (PGRN A9D) results in cytoplasmic missorting with extremely low expression. In contrast, two other progranulin mutations (PGRN P248L and R432C) are expressed as immature proteins but are inefficiently transported through and partially degraded within the secretory pathway, resulting in a significantly reduced secretion. Thus apparently all progranulin mutations cause reduced protein expression or secretion, although by different cellular mechanisms. To investigate a putative relationship between reduced expression of progranulin and TDP-43 relocalization and deposition, we down-regulated progranulin in human cell lines and in zebrafish. Upon reduction of progranulin, neither a major redistribution of TDP-43 nor proteolytic processing to disease-characterizing C-terminal fragments could be observed.


Asunto(s)
Demencia/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cuerpos de Inclusión Intranucleares/metabolismo , Mutación Missense/genética , Ubiquitina/inmunología , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Progranulinas , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Fracciones Subcelulares/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...