Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38888964

RESUMEN

The ß-secretase BACE1 is a central drug target for Alzheimer's disease. Clinically tested, BACE1-directed inhibitors also block the homologous protease BACE2. Yet, little is known about physiological BACE2 substrates and functions in vivo. Here, we identify BACE2 as the protease shedding the lymphangiogenic vascular endothelial growth factor receptor 3 (VEGFR3). Inactivation of BACE2, but not BACE1, inhibited shedding of VEGFR3 from primary human lymphatic endothelial cells (LECs) and reduced release of the shed, soluble VEGFR3 (sVEGFR3) ectodomain into the blood of mice, non-human primates and humans. Functionally, BACE2 inactivation increased full-length VEGFR3 and enhanced VEGFR3 signaling in LECs and also in vivo in zebrafish, where enhanced migration of LECs was observed. Thus, this study identifies BACE2 as a modulator of lymphangiogenic VEGFR3 signaling and demonstrates the utility of sVEGFR3 as a pharmacodynamic plasma marker for BACE2 activity in vivo, a prerequisite for developing BACE1-selective inhibitors for a safer prevention of Alzheimer's disease.

2.
Mol Neurodegener ; 19(1): 50, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902734

RESUMEN

BACKGROUND: The key pathological signature of ALS/ FTLD is the mis-localization of endogenous TDP-43 from the nucleus to the cytoplasm. However, TDP-43 gain of function in the cytoplasm is still poorly understood since TDP-43 animal models recapitulating mis-localization of endogenous TDP-43 from the nucleus to the cytoplasm are missing. METHODS: CRISPR/Cas9 technology was used to generate a zebrafish line (called CytoTDP), that mis-locates endogenous TDP-43 from the nucleus to the cytoplasm. Phenotypic characterization of motor neurons and the neuromuscular junction was performed by immunostaining, microglia were immunohistochemically localized by whole-mount tissue clearing and muscle ultrastructure was analyzed by scanning electron microscopy. Behavior was investigated by video tracking and quantitative analysis of swimming parameters. RNA sequencing was used to identify mis-regulated pathways with validation by molecular analysis. RESULTS: CytoTDP fish have early larval phenotypes resembling clinical features of ALS such as progressive motor defects, neurodegeneration and muscle atrophy. Taking advantage of zebrafish's embryonic development that solely relys on yolk usage until 5 days post fertilization, we demonstrated that microglia proliferation and activation in the hypothalamus is independent from food intake. By comparing CytoTDP to a previously generated TDP-43 knockout line, transcriptomic analyses revealed that mis-localization of endogenous TDP-43, rather than TDP-43 nuclear loss of function, leads to early onset metabolic dysfunction. CONCLUSIONS: The new TDP-43 model mimics the ALS/FTLD hallmark of progressive motor dysfunction. Our results suggest that functional deficits of the hypothalamus, the metabolic regulatory center, might be the primary cause of weight loss in ALS patients. Cytoplasmic gain of function of endogenous TDP-43 leads to metabolic dysfunction in vivo that are reminiscent of early ALS clinical non-motor metabolic alterations. Thus, the CytoTDP zebrafish model offers a unique opportunity to identify mis-regulated targets for therapeutic intervention early in disease progression.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Neuronas Motoras , Proteínas de Pez Cebra , Pez Cebra , Animales , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Animales Modificados Genéticamente , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología
3.
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.

4.
Nat Neurosci ; 25(12): 1608-1625, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36424432

RESUMEN

Decreasing the activation of pathology-activated microglia is crucial to prevent chronic inflammation and tissue scarring. In this study, we used a stab wound injury model in zebrafish and identified an injury-induced microglial state characterized by the accumulation of lipid droplets and TAR DNA-binding protein of 43 kDa (TDP-43)+ condensates. Granulin-mediated clearance of both lipid droplets and TDP-43+ condensates was necessary and sufficient to promote the return of microglia back to the basal state and achieve scarless regeneration. Moreover, in postmortem cortical brain tissues from patients with traumatic brain injury, the extent of microglial activation correlated with the accumulation of lipid droplets and TDP-43+ condensates. Together, our results reveal a mechanism required for restoring microglia to a nonactivated state after injury, which has potential for new therapeutic applications in humans.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Microglía , Humanos , Animales , Gotas Lipídicas , Pez Cebra , Proteínas de Unión al ADN , Regeneración
5.
Nat Med ; 22(7): 735-43, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27294876

RESUMEN

Immunomodulatory drugs (IMiDs), such as thalidomide and its derivatives lenalidomide and pomalidomide, are key treatment modalities for hematologic malignancies, particularly multiple myeloma (MM) and del(5q) myelodysplastic syndrome (MDS). Cereblon (CRBN), a substrate receptor of the CRL4 ubiquitin ligase complex, is the primary target by which IMiDs mediate anticancer and teratogenic effects. Here we identify a ubiquitin-independent physiological chaperone-like function of CRBN that promotes maturation of the basigin (BSG; also known as CD147) and solute carrier family 16 member 1 (SLC16A1; also known as MCT1) proteins. This process allows for the formation and activation of the CD147-MCT1 transmembrane complex, which promotes various biological functions, including angiogenesis, proliferation, invasion and lactate export. We found that IMiDs outcompete CRBN for binding to CD147 and MCT1, leading to destabilization of the CD147-MCT1 complex. Accordingly, IMiD-sensitive MM cells lose CD147 and MCT1 expression after being exposed to IMiDs, whereas IMiD-resistant cells retain their expression. Furthermore, del(5q) MDS cells have elevated CD147 expression, which is attenuated after IMiD treatment. Finally, we show that BSG (CD147) knockdown phenocopies the teratogenic effects of thalidomide exposure in zebrafish. These findings provide a common mechanistic framework to explain both the teratogenic and pleiotropic antitumor effects of IMiDs.


Asunto(s)
Basigina/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Factores Inmunológicos/farmacología , Inmunosupresores/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Péptido Hidrolasas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Teratogénesis/efectos de los fármacos , Talidomida/farmacología , Proteínas Adaptadoras Transductoras de Señales , Basigina/genética , Basigina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos , Lenalidomida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Teratogénesis/genética , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligasas
6.
Nat Neurosci ; 19(1): 102-10, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26656643

RESUMEN

The ability to form memories is a prerequisite for an organism's behavioral adaptation to environmental changes. At the molecular level, the acquisition and maintenance of memory requires changes in chromatin modifications. In an effort to unravel the epigenetic network underlying both short- and long-term memory, we examined chromatin modification changes in two distinct mouse brain regions, two cell types and three time points before and after contextual learning. We found that histone modifications predominantly changed during memory acquisition and correlated surprisingly little with changes in gene expression. Although long-lasting changes were almost exclusive to neurons, learning-related histone modification and DNA methylation changes also occurred in non-neuronal cell types, suggesting a functional role for non-neuronal cells in epigenetic learning. Finally, our data provide evidence for a molecular framework of memory acquisition and maintenance, wherein DNA methylation could alter the expression and splicing of genes involved in functional plasticity and synaptic wiring.


Asunto(s)
Conducta Animal/fisiología , Región CA1 Hipocampal/metabolismo , Cromatina/química , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Expresión Génica/fisiología , Giro del Cíngulo/metabolismo , Histonas/metabolismo , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/fisiología , Plasticidad Neuronal/fisiología , Animales , Condicionamiento Psicológico , Metilación de ADN/genética , Epigénesis Genética/genética , Miedo , Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/genética
7.
J Neurosci ; 33(18): 7856-69, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23637177

RESUMEN

Proteolytic shedding of cell surface proteins generates paracrine signals involved in numerous signaling pathways. Neuregulin 1 (NRG1) type III is involved in myelination of the peripheral nervous system, for which it requires proteolytic activation by proteases of the ADAM family and BACE1. These proteases are major therapeutic targets for the prevention of Alzheimer's disease because they are also involved in the proteolytic generation of the neurotoxic amyloid ß-peptide. Identification and functional investigation of their physiological substrates is therefore of greatest importance in preventing unwanted side effects. Here we investigated proteolytic processing of NRG1 type III and demonstrate that the ectodomain can be cleaved by three different sheddases, namely ADAM10, ADAM17, and BACE1. Surprisingly, we not only found cleavage by ADAM10, ADAM17, and BACE1 C-terminal to the epidermal growth factor (EGF)-like domain, which is believed to play a pivotal role in signaling, but also additional cleavage sites for ADAM17 and BACE1 N-terminal to that domain. Proteolytic processing at N- and C-terminal sites of the EGF-like domain results in the secretion of this domain from NRG1 type III. The soluble EGF-like domain is functionally active and stimulates ErbB3 signaling in tissue culture assays. Moreover, the soluble EGF-like domain is capable of rescuing hypomyelination in a zebrafish mutant lacking BACE1. Our data suggest that NRG1 type III-dependent myelination is not only controlled by membrane-retained NRG1 type III, but also in a paracrine manner via proteolytic liberation of the EGF-like domain.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Neurregulinas/metabolismo , Comunicación Paracrina/fisiología , Proteína ADAM17 , Animales , Membrana Celular/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , Embrión de Mamíferos , Factor de Crecimiento Epidérmico/análogos & derivados , Factor de Crecimiento Epidérmico/química , Humanos , Inmunoprecipitación , Neurregulinas/genética , Neuronas , Fosforilación , Proteolisis , ARN Mensajero/administración & dosificación , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Células de Schwann , Transfección , Pez Cebra
8.
Proteomics ; 13(9): 1519-27, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23457027

RESUMEN

The aspartyl protease BACE1 cleaves neuregulin 1 and is involved in myelination and is a candidate drug target for Alzheimer's disease, where it acts as the ß-secretase cleaving the amyloid precursor protein. However, little is known about other substrates in vivo. Here, we provide a proteomic workflow for BACE1 substrate identification from whole brains, combining filter-aided sample preparation, strong-anion exchange fractionation, and label-free quantification. We used bace1-deficient zebrafish and quantified differences in protein levels between wild-type and bace1 -/- zebrafish brains. Over 4500 proteins were identified with at least two unique peptides and quantified in both wild-type and bace1 -/- zebrafish brains. The majority of zebrafish membrane proteins did not show altered protein levels, indicating that Bace1 has a restricted substrate specificity. Twenty-four membrane proteins accumulated in the bace1 -/- brains and thus represent candidate Bace1 substrates. They include several known BACE1 substrates, such as the zebrafish homologs of amyloid precursor protein and the cell adhesion protein L1, which validate the proteomic workflow. Additionally, several candidate substrates with a function in neurite outgrowth and axon guidance, such as plexin A3 and glypican-1 were identified, pointing to a function of Bace1 in neurodevelopment. Taken together, our study provides the first proteomic analysis of knock-out zebrafish tissue and demonstrates that combining gene knock-out models in zebrafish with quantitative proteomics is a powerful approach to address biomedical questions.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Ácido Aspártico Endopeptidasas/genética , Encéfalo/metabolismo , Proteínas de la Membrana/análisis , Proteínas de Pez Cebra/análisis , Pez Cebra/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Ácido Aspártico Endopeptidasas/metabolismo , Fraccionamiento Químico/métodos , Glipicanos/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteoma/genética , Proteoma/metabolismo , Receptores de Superficie Celular/metabolismo , Espectrometría de Masas en Tándem , Flujo de Trabajo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
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
10.
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
11.
PLoS One ; 5(7): e11783, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20689587

RESUMEN

BACKGROUND: Mutations in the gene encoding the E3 ubiquitin ligase parkin (PARK2) are responsible for the majority of autosomal recessive parkinsonism. Similarly to other knockout mouse models of PD-associated genes, parkin knockout mice do not show a substantial neuropathological or behavioral phenotype, while loss of parkin in Drosophila melanogaster leads to a severe phenotype, including reduced lifespan, apoptotic flight muscle degeneration and male sterility. In order to study the function of parkin in more detail and to address possible differences in its role in different species, we chose Danio rerio as a different vertebrate model system. METHODOLOGY/PRINCIPAL FINDINGS: We first cloned zebrafish parkin to compare its biochemical and functional aspects with that of human parkin. By using an antisense knockdown strategy we generated a zebrafish model of parkin deficiency (knockdown efficiency between 50% and 60%) and found that the transient knockdown of parkin does not cause morphological or behavioral alterations. Specifically, we did not observe a loss of dopaminergic neurons in parkin-deficient zebrafish. In addition, we established transgenic zebrafish lines stably expressing parkin by using a Gal4/UAS-based bidirectional expression system. While parkin-deficient zebrafish are more vulnerable to proteotoxicity, increased parkin expression protected transgenic zebrafish from cell death induced by proteotoxic stress. CONCLUSIONS/SIGNIFICANCE: Similarly to human parkin, zebrafish parkin is a stress-responsive protein which protects cells from stress-induced cell death. Our transgenic zebrafish model is a novel tool to characterize the protective capacity of parkin in vivo.


Asunto(s)
Dopamina/metabolismo , Neuronas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Línea Celular , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Células HeLa , Humanos , Hibridación in Situ , Masculino , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/fisiología , Reacción en Cadena de la Polimerasa , Ubiquitina-Proteína Ligasas/genética , Pez Cebra/genética
12.
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
13.
Dev Biol ; 327(1): 177-90, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19133254

RESUMEN

Epimorphic regeneration is a unique and complex instance of postembryonic growth observed in certain metazoans that is usually triggered by severe injury [Akimenko et al., 2003; Alvarado and Tsonis, 2006; Brockes, 1997; Endo et al., 2004]. Cell division and migration are two fundamental biological processes required for supplying replacement cells during regeneration [Endo et al., 2004; Slack, 2007]. However, the connection between the early stimuli generated after injury and the signals regulating proliferation and migration during regeneration remain largely unknown. Here we show that the oncogenes ErbB2 and ErbB3, two members of the EGFR family, are essential for mounting a successful regeneration response in vertebrates. Importantly, amputation-induced progenitor proliferation and migration are significantly reduced upon genetic and/or chemical modulation of ErbB function. Moreover, we also found that NRG1 and PI3K functionally interact with ErbB2 and ErbB3 during regeneration and interfering with their function also abrogates the capacity of progenitor cells to regenerate lost structures upon amputation. Our findings suggest that ErbB, PI3K and NRG1 are components of a permissive switch for migration and proliferation continuously acting across the amputated fin from early stages of vertebrate regeneration onwards that regulate the expression of the transcription factors lef1 and msxB.


Asunto(s)
Amputación Quirúrgica , Receptor ErbB-2/fisiología , Receptor ErbB-3/fisiología , Regeneración , Células Madre/fisiología , Animales , Movimiento Celular , Proliferación Celular , Proteínas de Homeodominio/genética , Neurregulina-1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Células Madre/citología , Factores de Transcripción/genética , Vertebrados , Pez Cebra , Proteínas de Pez Cebra/genética
14.
BMC Genomics ; 8: 11, 2007 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-17212827

RESUMEN

BACKGROUND: Large-scale mutagenesis screens in the zebrafish employing the mutagen ENU have isolated several hundred mutant loci that represent putative developmental control genes. In order to realize the potential of such screens, systematic genetic mapping of the mutations is necessary. Here we report on a large-scale effort to map the mutations generated in mutagenesis screening at the Max Planck Institute for Developmental Biology by genome scanning with microsatellite markers. RESULTS: We have selected a set of microsatellite markers and developed methods and scoring criteria suitable for efficient, high-throughput genome scanning. We have used these methods to successfully obtain a rough map position for 319 mutant loci from the Tübingen I mutagenesis screen and subsequent screening of the mutant collection. For 277 of these the corresponding gene is not yet identified. Mapping was successful for 80 % of the tested loci. By comparing 21 mutation and gene positions of cloned mutations we have validated the correctness of our linkage group assignments and estimated the standard error of our map positions to be approximately 6 cM. CONCLUSION: By obtaining rough map positions for over 300 zebrafish loci with developmental phenotypes, we have generated a dataset that will be useful not only for cloning of the affected genes, but also to suggest allelism of mutations with similar phenotypes that will be identified in future screens. Furthermore this work validates the usefulness of our methodology for rapid, systematic and inexpensive microsatellite mapping of zebrafish mutations.


Asunto(s)
Mapeo Cromosómico , Repeticiones de Microsatélite , Mutación , Pez Cebra/embriología , Pez Cebra/genética , Animales , Femenino , Genoma , Masculino , Mutagénesis , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...