Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Antibiot (Tokyo) ; 77(4): 245-256, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38238588

RESUMEN

Tunicamycins (TUN) are well-defined, Streptomyces-derived natural products that inhibit protein N-glycosylation in eukaryotes, and by a conserved mechanism also block bacterial cell wall biosynthesis. TUN inhibits the polyprenylphosphate-N-acetyl-hexosamine-1-phospho-transferases (PNPT), an essential family of enzymes found in both bacteria and eukaryotes. We have previously published the development of chemically modified TUN, called TunR1 and TunR2, that have considerably reduced activity on eukaryotes but that retain the potent antibacterial properties. A mechanism for this reduced toxicity has also been reported. TunR1 and TunR2 have been tested against mammalian cell lines in culture and against live insect cells but, until now, no in vivo evaluation has been undertaken for vertebrates. In the current work, TUN, TunR1, and TunR2 are investigated for their relative toxicity and antimycobacterial activity in zebrafish using a well-established Mycobacterium marinum (M. marinum) infection system, a model for studying human Mycobacterium tuberculosis infections. We also report the relative ability to activate the unfolded protein response (UPR), the known mechanism for the eukaryotic toxicity observed with TUN treatment. Importantly, TunR1 and TunR2 retained their antimicrobial properties, as evidenced by a reduction in M. marinum bacterial burden, compared to DMSO-treated zebrafish. In summary, findings from this study highlight the characteristics of recently developed TUN derivatives, mainly TunR2, and its potential for use as a novel anti-bacterial agent for veterinary and potential medical purposes.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Mycobacterium marinum , Tunicamicina , Animales , Humanos , Antibacterianos/farmacología , Mamíferos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/fisiología , Tunicamicina/química , Tunicamicina/análogos & derivados , Pez Cebra/microbiología , Fosfotransferasas/química
2.
Arterioscler Thromb Vasc Biol ; 43(7): e231-e237, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37128914

RESUMEN

BACKGROUND: The goal of this study was to identify and characterize cell-cell interactions that facilitate endothelial tip cell fusion downstream of BMP (bone morphogenic protein)-mediated venous plexus formation. METHODS: High resolution and time-lapse imaging of transgenic reporter lines and loss-of-function studies were carried out to study the involvement of mesenchymal stromal cells during venous angiogenesis. RESULTS: BMP-responsive stromal cells facilitate timely and precise fusion of venous tip cells during developmental angiogenesis. CONCLUSIONS: Stromal cells are required for anastomosis of venous tip cells in the embryonic caudal hematopoietic tissue.


Asunto(s)
Proteínas Morfogenéticas Óseas , Células Madre Mesenquimatosas , Animales , Fusión Celular , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales Modificados Genéticamente , Comunicación Celular , Células del Estroma/metabolismo
3.
Invest Ophthalmol Vis Sci ; 62(15): 13, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34913948

RESUMEN

Purpose: Proper refractive development of the eye, termed emmetropization, is critical for focused vision and is impacted by both genetic determinants and several visual environment factors. Improper emmetropization caused by genetic variants can lead to congenital hyperopia, which is characterized by small eyes and relatively short ocular axial length. To date, variants in only four genes have been firmly associated with human hyperopia, one of which is MFRP. Zebrafish mfrp mutants also have hyperopia and, similar to reports in mice, exhibit increased macrophage recruitment to the retina. The goal of this research was to examine the effects of macrophage ablation on emmetropization and mfrp-related hyperopia. Methods: We utilized a chemically inducible, cell-specific ablation system to deplete macrophages in both wild-type and mfrp mutant zebrafish. Spectral-domain optical coherence tomography was then used to measure components of the eye and determine relative refractive state. Histology, immunohistochemistry, and transmission electron microscopy were used to further study the eyes. Results: Although macrophage ablation does not cause significant changes to the relative refractive state of wild-type zebrafish, macrophage ablation in mfrp mutants significantly exacerbates their hyperopic phenotype, resulting in a relative refractive error 1.3 times higher than that of non-ablated mfrp siblings. Conclusions: Genetic inactivation of mfrp leads to hyperopia, as well as abnormal accumulation of macrophages in the retina. Ablation of the mpeg1-positive macrophage population exacerbates the hyperopia, suggesting that macrophages may be recruited in an effort help preserve emmetropization and ameliorate hyperopia.


Asunto(s)
Proteínas del Ojo/genética , Hiperopía/fisiopatología , Macrófagos/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Antiinfecciosos/farmacología , Apoptosis , Proliferación Celular , Colágeno/metabolismo , Colágeno/ultraestructura , Emetropía/fisiología , Hiperopía/diagnóstico por imagen , Hiperopía/genética , Inmunohistoquímica , Metronidazol/farmacología , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Fenotipo , Refracción Ocular , Esclerótica/metabolismo , Esclerótica/ultraestructura , Tomografía de Coherencia Óptica , Pez Cebra
4.
Mol Biol Cell ; 32(5): 391-401, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33439675

RESUMEN

Coat protein complex II (COPII) factors mediate cargo export from the endoplasmic reticulum (ER), but bulky collagens and lipoproteins are too large for traditional COPII vesicles. Mammalian CTAGE5 and TANGO1 have been well characterized individually as specialized cargo receptors at the ER that function with COPII coats to facilitate trafficking of bulky cargoes. Here, we present a genetic interaction study in zebrafish of deletions in ctage5, tango1, or both to investigate their distinct and complementary potential functions. We found that Ctage5 and Tango1 have different roles related to organogenesis, collagen versus lipoprotein trafficking, stress-pathway activation, and survival. While disruption of both ctage5 and tango1 compounded phenotype severity, mutation of either factor alone revealed novel tissue-specific defects in the building of heart, muscle, lens, and intestine, in addition to previously described roles in the development of neural and cartilage tissues. Together, our results demonstrate that Ctage5 and Tango1 have overlapping functions, but also suggest divergent roles in tissue development and homeostasis.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Proteínas de Neoplasias/genética , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Homeostasis , Proteínas de Neoplasias/metabolismo , Transporte de Proteínas/fisiología , Proteínas de Transporte Vesicular/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Drug Discov Today ; 26(8): 1790-1793, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33358701

RESUMEN

Capillary malformation-arteriovenous malformation (CM-AVM) syndrome is a class of capillary anomalies that are associated with arteriovenous malformations and arteriovenous fistulas, which carry a risk of hemorrhages. There are no broadly effective pharmacological therapies currently available. Most CM-AVMs are associated with a loss of RASA1, resulting in constitutive activation of RAS signaling. However, protein interaction analysis revealed that RASA1 forms a complex with Rho GTPase-activating protein (RhoGAP), a negative regulator of RhoA signaling. Herein, we propose that loss of RASA1 function results in constitutive activation of RhoA signaling in endothelial cells, resulting in enhanced vascular permeability. Therefore, strategies aimed at curtailing RhoA activity should be tested as an adjunctive therapeutic approach in cell culture studies and animal models of RASA1 deficiency.


Asunto(s)
Malformaciones Arteriovenosas/fisiopatología , Capilares/anomalías , Mancha Vino de Oporto/fisiopatología , Proteína Activadora de GTPasa p120/genética , Proteína de Unión al GTP rhoA/genética , Animales , Malformaciones Arteriovenosas/tratamiento farmacológico , Malformaciones Arteriovenosas/genética , Capilares/fisiopatología , Permeabilidad Capilar/fisiología , Células Endoteliales/citología , Humanos , Mutación , Mancha Vino de Oporto/tratamiento farmacológico , Mancha Vino de Oporto/genética , Transducción de Señal/fisiología
6.
Development ; 147(22)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33060129

RESUMEN

Adherens junction remodeling regulated by apical polarity proteins constitutes a major driving force for tissue morphogenesis, although the precise mechanism remains inconclusive. Here, we report that, in zebrafish, the Crumbs complex component MPP5a interacts with small GTPase Rab11 in Golgi to transport cadherin and Crumbs components synergistically to the apical domain, thus establishing apical epithelial polarity and adherens junctions. In contrast, Par complex recruited by MPP5a is incapable of interacting with Rab11 but might assemble cytoskeleton to facilitate cadherin exocytosis. In accordance, dysfunction of MPP5a induces an invasive migration of epithelial cells. This adherens junction remodeling pattern is frequently observed in zebrafish lens epithelial cells and neuroepithelial cells. The data identify an unrecognized MPP5a-Rab11 complex and describe its essential role in guiding apical polarization and zonula adherens formation in epithelial cells.


Asunto(s)
Uniones Adherentes/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Guanilato Ciclasa/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Proteínas de Unión al GTP rab/metabolismo , Uniones Adherentes/genética , Animales , Cadherinas/genética , Cadherinas/metabolismo , Células Epiteliales , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Guanilato Ciclasa/genética , Transporte de Proteínas/fisiología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Unión al GTP rab/genética
7.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32439761

RESUMEN

The development of the biliary system is a complex yet poorly understood process, with relevance to multiple diseases, including biliary atresia, choledochal cysts and gallbladder agenesis. We present here a crucial role for Hippo-Yap/Taz signaling in this context. Analysis of sav1 mutant zebrafish revealed dysplastic morphology and expansion of both intrahepatic and extrahepatic biliary cells, and ultimately larval lethality. Biliary dysgenesis, but not larval lethality, is driven primarily by Yap signaling. Re-expression of Sav1 protein in sav1-/- hepatocytes is able to overcome these initial deficits and allows sav1-/- fish to survive, suggesting cell non-autonomous signaling from hepatocytes. Examination of sav1-/- rescued adults reveals loss of gallbladder and formation of dysplastic cell masses expressing biliary markers, suggesting roles for Hippo signaling in extrahepatic biliary carcinomas. Deletion of stk3 revealed that the phenotypes observed in sav1 mutant fish function primarily through canonical Hippo signaling and supports a role for phosphatase PP2A, but also suggests Sav1 has functions in addition to facilitating Stk3 activity. Overall, this study defines a role for Hippo-Yap signaling in the maintenance of both intra- and extrahepatic biliary ducts.


Asunto(s)
Sistema Biliar/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Aciltransferasas , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Sistema Biliar/anatomía & histología , Sistema Biliar/crecimiento & desarrollo , Sistemas CRISPR-Cas/genética , Hidrolasas de Éster Carboxílico/metabolismo , Vesícula Biliar/anatomía & histología , Vesícula Biliar/crecimiento & desarrollo , Vesícula Biliar/metabolismo , Larva/crecimiento & desarrollo , Larva/metabolismo , Hígado/anatomía & histología , Hígado/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Serina-Treonina Quinasa 3 , Transducción de Señal , Transactivadores/genética , Factores de Transcripción/genética , Proteínas Señalizadoras YAP , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
8.
Cells ; 8(9)2019 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-31450674

RESUMEN

The cellular signaling pathways underlying peripheral nerve sheath tumor (PNST) formation are poorly understood. Hippo signaling has been recently implicated in the biology of various cancers, and is thought to function downstream of mutations in the known PNST driver, NF2. Utilizing CRISPR-Cas9 gene editing, we targeted the canonical Hippo signaling kinase Lats2. We show that, while germline deletion leads to early lethality, targeted somatic mutations of zebrafish lats2 leads to peripheral nerve sheath tumor formation. These peripheral nerve sheath tumors exhibit high levels of Hippo effectors Yap and Taz, suggesting that dysregulation of these transcriptional co-factors drives PNST formation in this model. These data indicate that somatic lats2 deletion in zebrafish can serve as a powerful experimental platform to probe the mechanisms of PNST formation and progression.


Asunto(s)
Mutación , Neoplasias Experimentales/patología , Neoplasias de la Vaina del Nervio/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Pez Cebra/genética , Animales , Sistemas CRISPR-Cas , Proliferación Celular , Edición Génica , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Serina-Treonina Quinasa 3 , Transducción de Señal , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/metabolismo
9.
Cells ; 8(5)2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31137701

RESUMEN

Hippo signaling controls cellular processes that ultimately impact organogenesis and homeostasis. Consequently, disease states including cancer can emerge when signaling is deregulated. The major pathway transducers Yap and Taz require cofactors to impart transcriptional control over target genes. Research into Yap/Taz-mediated epigenetic modifications has revealed their association with chromatin-remodeling complex proteins as a means of altering chromatin structure, therefore affecting accessibility and activity of target genes. Specifically, Yap/Taz have been found to associate with factors of the GAGA, Ncoa6, Mediator, Switch/sucrose nonfermentable (SWI/SNF), and Nucleosome Remodeling and Deacetylase (NuRD) chromatin-remodeling complexes to alter the accessibility of target genes. This review highlights the different mechanisms by which Yap/Taz collaborate with other factors to modify DNA packing at specific loci to either activate or repress target gene transcription.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Genes de Cambio/fisiología , Vía de Señalización Hippo , Histona Metiltransferasas/metabolismo , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
10.
Curr Biol ; 29(2): 242-255.e6, 2019 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-30595521

RESUMEN

The Hippo pathway plays major roles in development, regeneration, and cancer. Its activity is tightly regulated by both diffusible chemical ligands and mechanical stimuli. The pathway consists of a series of kinases that can control the sub-cellular localization and stability of YAP or TAZ, homologous transcriptional co-factors. Caveolae, small (60-100 nm) bulb-like invaginations of the plasma membrane, are comprised predominantly of caveolin and cavin proteins and can respond to mechanical stimuli. Here, we show that YAP/TAZ, the major transcriptional mediators of the Hippo pathway, are critical for expression of caveolae components and therefore caveolae formation in both mammalian cells and zebrafish. In essence, without YAP/TAZ, the cell loses an entire organelle. CAVEOLIN1 and CAVIN1, the two essential caveolar genes, are direct target genes of YAP/TAZ, regulated via TEA domain (TEAD) transcription factors. Notably, YAP/TAZ become nuclear enriched and facilitate target gene transcription in cells with diminished levels of caveolae. Furthermore, caveolar-mediated shear stress response activates YAP/TAZ. These data link caveolae to Hippo signaling in the context of cellular responses to mechanical stimuli and suggest activity-based feedback regulation between components of caveolae and the outputs of the Hippo pathway.


Asunto(s)
Caveolas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/fisiología , Proteínas de Pez Cebra/genética , Pez Cebra/fisiología , Animales , Células HEK293 , Vía de Señalización Hippo , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Serina-Treonina Quinasa 3 , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
Cardiovasc Res ; 115(3): 570-577, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295714

RESUMEN

AIMS: The Hippo signalling pathway regulates multiple cellular processes during organ development and maintenance by modulating activity of the transcriptional cofactor Yap. Core components of this pathway are required for neonatal mouse heart regeneration, however, investigations to date have typically focused on expression and activity in cardiomyocytes. Due to the regenerative capacity of zebrafish and the fact that global loss of Yap is not fully embryonic lethal in zebrafish, we leveraged a yap null mutant to investigate the impact of constitutive Yap deletion during zebrafish heart regeneration. METHODS AND RESULTS: Following cryoinjury in adult hearts, myocyte proliferation was not decreased in yap mutants, contrary to expectations based on mouse data. Experiments in larval zebrafish (Danio rerio) revealed that deletion of either Yap or Taz had a modest effect on heart growth, reducing gross organ size, while their combined deletion was synergistic; thus, Yap and Taz share some overlapping roles in zebrafish heart development. Surprisingly, adult yap mutants exhibited decreased collagen composition at 7 days post-injury, suggesting a critical role for Yap in scar formation during heart regeneration. siRNA-mediated Yap knockdown in primary rat (Rattus norvegicus) cardiac cells revealed a fibroblast-specific role for Yap in controlling the expression of cytoskeletal and myofibroblast activation genes, as well as pro-inflammatory cyto/chemokines. Corroborating these RNAseq data, we observed increased macrophage infiltration in the scars of yap mutants at 7 days post-injury. CONCLUSION: These results suggest that Yap deletion has minimal effect on myocyte proliferation in adults, but significantly influences scar formation and immune cell infiltration during zebrafish heart regeneration. Collectively, these data suggest an unexpected role for Yap in matrix formation and macrophage recruitment during heart regeneration.


Asunto(s)
Proliferación Celular , Cicatriz/metabolismo , Lesiones Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Regeneración , Transactivadores/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Cultivadas , Cicatriz/genética , Cicatriz/patología , Cicatriz/fisiopatología , Frío , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Lesiones Cardíacas/genética , Lesiones Cardíacas/patología , Lesiones Cardíacas/fisiopatología , Macrófagos/metabolismo , Macrófagos/patología , Miocitos Cardíacos/patología , Ratas , Transducción de Señal , Transactivadores/genética , Remodelación Ventricular , Proteínas Señalizadoras YAP , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
12.
Mol Cell ; 71(2): 216-228.e7, 2018 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-30029002

RESUMEN

The polyglutamine (polyQ) diseases are a group of nine neurodegenerative diseases caused by the expansion of a polyQ tract that results in protein aggregation. Unlike other model organisms, Dictyostelium discoideum is a proteostatic outlier, naturally encoding long polyQ tracts yet resistant to polyQ aggregation. Here we identify serine-rich chaperone protein 1 (SRCP1) as a molecular chaperone that is necessary and sufficient to suppress polyQ aggregation. SRCP1 inhibits aggregation of polyQ-expanded proteins, allowing for their degradation via the proteasome, where SRCP1 is also degraded. SRCP1's C-terminal domain is essential for its activity in cells, and peptides that mimic this domain suppress polyQ aggregation in vitro. Together our results identify a novel type of molecular chaperone and reveal how nature has dealt with the problem of polyQ aggregation.


Asunto(s)
Chaperonas Moleculares/metabolismo , Péptidos/metabolismo , Línea Celular , Dictyostelium/metabolismo , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Serina/metabolismo , Ubiquitina/metabolismo
13.
Hum Mol Genet ; 27(10): 1675-1695, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29506241

RESUMEN

The PITX2 (paired-like homeodomain 2) gene encodes a bicoid-like homeodomain transcription factor linked with several human disorders. The main associated congenital phenotype is Axenfeld-Rieger syndrome, type 1, an autosomal dominant condition characterized by variable defects in the anterior segment of the eye, an increased risk of glaucoma, craniofacial dysmorphism and dental and umbilical anomalies; in addition to this, one report implicated PITX2 in ring dermoid of the cornea and a few others described cardiac phenotypes. We report three novel PITX2 mutations-c.271C > T, p.(Arg91Trp); c.259T > C, p.(Phe87Leu); and c.356delA, p.(Gln119Argfs*36)-identified in independent families with typical Axenfeld-Rieger syndrome characteristics and some unusual features such as corneal guttata, Wolf-Parkinson-White syndrome, and hyperextensibility. To gain further insight into the diverse roles of PITX2/pitx2 in vertebrate development, we generated various genetic lesions in the pitx2 gene via TALEN-mediated genome editing. Affected homozygous zebrafish demonstrated congenital defects consistent with the range of PITX2-associated human phenotypes: abnormal development of the cornea, iris and iridocorneal angle; corneal dermoids; and craniofacial dysmorphism. In addition, via comparison of pitx2M64* and wild-type embryonic ocular transcriptomes we defined molecular changes associated with pitx2 deficiency, thereby implicating processes potentially underlying disease pathology. This analysis identified numerous affected factors including several members of the Wnt pathway and collagen types I and V gene families. These data further support the link between PITX2 and the WNT pathway and suggest a new role in regulation of collagen gene expression during development.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Anomalías del Ojo/genética , Enfermedades Hereditarias del Ojo/genética , Glaucoma/genética , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Síndrome de Wolff-Parkinson-White/genética , Proteínas de Pez Cebra/genética , Animales , Segmento Anterior del Ojo/fisiopatología , Colágeno Tipo I/genética , Colágeno Tipo V/genética , Anomalías del Ojo/fisiopatología , Enfermedades Hereditarias del Ojo/fisiopatología , Edición Génica , Regulación del Desarrollo de la Expresión Génica , Glaucoma/fisiopatología , Humanos , Mutación , Linaje , Factores de Transcripción/deficiencia , Vía de Señalización Wnt , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteína del Homeodomínio PITX2
14.
Invest Ophthalmol Vis Sci ; 57(15): 6805-6814, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28002843

RESUMEN

Purpose: Mutations in membrane frizzled-related protein (MFRP) are associated with nanophthalmia, hyperopia, foveoschisis, irregular patches of RPE atrophy, and optic disc drusen in humans. Mouse mfrp mutants show retinal degeneration but no change in eye size or refractive state. The goal of this work was to generate zebrafish mutants to investigate the loss of Mfrp on eye size and refractive state, and to characterize other phenotypes observed. Methods: Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 methods were used to generate multiple frameshift mutations in zebrafish mfrp causing premature translational stops in Mfrp. Spectral-domain optical coherence tomography (SD-OCT) was used to measure eye metrics and refractive state, and immunohistochemistry was used to study adult eyes. Gene expression levels were measured using quantitative PCR. Results: Zebrafish Mfrp was shown to localize to apical and basal regions of RPE cells, as well as the ciliary marginal zone. Loss of Mfrp in mutant zebrafish was verified histologically. Zebrafish eyes that were mfrp mutant showed reduced axial length causing hyperopia, RPE folding, and macrophages were observed subretinally. Visual acuity was reduced in mfrp mutant animals. Conclusions: Mutation of zebrafish mfrp results in hyperopia with subretinal macrophage infiltration, phenocopying aspects of human and mouse Mfrp deficiency. These mutant zebrafish will be useful in studying the onset and progression of Mfrp-related nanophthalmia, the cues that initiate the recruitment of macrophages, and the mechanisms of Mfrp function.


Asunto(s)
Glicoproteínas/genética , Hiperopía/genética , Macrófagos/patología , Microftalmía/genética , Mutación , Degeneración Retiniana/genética , Epitelio Pigmentado de la Retina/patología , Animales , ADN/genética , Análisis Mutacional de ADN , Glicoproteínas/metabolismo , Humanos , Hiperopía/metabolismo , Hiperopía/patología , Péptidos y Proteínas de Señalización Intracelular , Microftalmía/metabolismo , Microftalmía/patología , Fenotipo , Reacción en Cadena de la Polimerasa , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/metabolismo , Tomografía de Coherencia Óptica , Pez Cebra
15.
Development ; 142(17): 3021-32, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26209646

RESUMEN

The optic vesicle comprises a pool of bi-potential progenitor cells from which the retinal pigment epithelium (RPE) and neural retina fates segregate during ocular morphogenesis. Several transcription factors and signaling pathways have been shown to be important for RPE maintenance and differentiation, but an understanding of the initial fate specification and determination of this ocular cell type is lacking. We show that Yap/Taz-Tead activity is necessary and sufficient for optic vesicle progenitors to adopt RPE identity in zebrafish. A Tead-responsive transgene is expressed within the domain of the optic cup from which RPE arises, and Yap immunoreactivity localizes to the nuclei of prospective RPE cells. yap (yap1) mutants lack a subset of RPE cells and/or exhibit coloboma. Loss of RPE in yap mutants is exacerbated in combination with taz (wwtr1) mutant alleles such that, when Yap and Taz are both absent, optic vesicle progenitor cells completely lose their ability to form RPE. The mechanism of Yap-dependent RPE cell type determination is reliant on both nuclear localization of Yap and interaction with a Tead co-factor. In contrast to loss of Yap and Taz, overexpression of either protein within optic vesicle progenitors leads to ectopic pigmentation in a dosage-dependent manner. Overall, this study identifies Yap and Taz as key early regulators of RPE genesis and provides a mechanistic framework for understanding the congenital ocular defects of Sveinsson's chorioretinal atrophy and congenital retinal coloboma.


Asunto(s)
Linaje de la Célula , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Epitelio Pigmentado de la Retina/citología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Alelos , Animales , Apoptosis/genética , Núcleo Celular/metabolismo , Proliferación Celular , Coloboma/patología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Morfogénesis/genética , Mutación , Fenotipo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Epitelio Pigmentado de la Retina/trasplante , Transducción de Señal/genética , Factores de Transcripción de Dominio TEA , Transactivadores/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Transgenes , Regulación hacia Arriba , Proteínas Señalizadoras YAP , Pez Cebra/genética , Proteínas de Pez Cebra/genética
16.
Nat Med ; 20(7): 732-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24973920

RESUMEN

Hedgehog signaling drives oncogenesis in several cancers, and strategies targeting this pathway have been developed, most notably through inhibition of Smoothened (SMO). However, resistance to Smoothened inhibitors occurs by genetic changes of Smoothened or other downstream Hedgehog components. Here we overcome these resistance mechanisms by modulating GLI transcription through inhibition of bromo and extra C-terminal (BET) bromodomain proteins. We show that BRD4 and other BET bromodomain proteins regulate GLI transcription downstream of SMO and suppressor of fused (SUFU), and chromatin immunoprecipitation studies reveal that BRD4 directly occupies GLI1 and GLI2 promoters, with a substantial decrease in engagement of these sites after treatment with JQ1, a small-molecule inhibitor targeting BRD4. Globally, genes associated with medulloblastoma-specific GLI1 binding sites are downregulated in response to JQ1 treatment, supporting direct regulation of GLI activity by BRD4. Notably, patient- and GEMM (genetically engineered mouse model)-derived Hedgehog-driven tumors (basal cell carcinoma, medulloblastoma and atypical teratoid rhabdoid tumor) respond to JQ1 even when harboring genetic lesions rendering them resistant to Smoothened antagonists. Altogether, our results reveal BET proteins as critical regulators of Hedgehog pathway transcriptional output and nominate BET bromodomain inhibitors as a strategy for treating Hedgehog-driven tumors with emerged or a priori resistance to Smoothened antagonists.


Asunto(s)
Epigénesis Genética , Proteínas Hedgehog/genética , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Animales , Azepinas/farmacología , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Ligandos , Ratones , Neoplasias Experimentales/patología , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Triazoles/farmacología , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
17.
Hum Mol Genet ; 20(20): 4041-55, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21816947

RESUMEN

Ciliopathies are a genetically and phenotypically heterogeneous group of human developmental disorders whose root cause is the absence or dysfunction of primary cilia. Joubert syndrome is characterized by a distinctive hindbrain malformation variably associated with retinal dystrophy and cystic kidney disease. Mutations in CC2D2A are found in ∼10% of patients with Joubert syndrome. Here we describe the retinal phenotype of cc2d2a mutant zebrafish consisting of disorganized rod and cone photoreceptor outer segments resulting in abnormal visual function as measured by electroretinogram. Our analysis reveals trafficking defects in mutant photoreceptors affecting transmembrane outer segment proteins (opsins) and striking accumulation of vesicles, suggesting a role for Cc2d2a in vesicle trafficking and fusion. This is further supported by mislocalization of Rab8, a key regulator of opsin carrier vesicle trafficking, in cc2d2a mutant photoreceptors and by enhancement of the cc2d2a retinal and kidney phenotypes with partial knockdown of rab8. We demonstrate that Cc2d2a localizes to the connecting cilium in photoreceptors and to the transition zone in other ciliated cell types and that cilia are present in these cells in cc2d2a mutants, arguing against a primary function for Cc2d2a in ciliogenesis. Our data support a model where Cc2d2a, localized at the photoreceptor connecting cilium/transition zone, facilitates protein transport through a role in Rab8-dependent vesicle trafficking and fusion.


Asunto(s)
Cilios/genética , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Vesículas Transportadoras/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Proteínas de Unión al GTP rab/genética , Animales , Animales Modificados Genéticamente , Cilios/metabolismo , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Unión Proteica , Transporte de Proteínas , Vesículas Transportadoras/ultraestructura , Proteínas de Transporte Vesicular/metabolismo , Visión Ocular/genética , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Unión al GTP rab/metabolismo
18.
Dev Dyn ; 240(3): 712-22, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21337469

RESUMEN

Bone morphogenic protein (BMP) signaling is fundamental to development, injury response, and homeostasis. We have developed transgenic zebrafish that report Smad-mediated BMP signaling in embryos and adults. These lines express either enhanced green fluorescent protein (eGFP), destabilized eGFP, or destabilized Kusabira Orange 2 (KO2) under the well-characterized BMP Response Element (BRE). These fluorescent proteins were found to be expressed dynamically in regions of known BMP signaling including the developing tail bud, hematopoietic lineage, dorsal eye, brain structures, heart, jaw, fins, and somites, as well as other tissues. Responsiveness to changes in BMP signaling was confirmed by observing fluorescence after activation in an hsp70:bmp2b transgenic background or by inhibition in an hsp70:nog3 background. We further demonstrated faithful reportage by the BRE transgenic lines following chemical repression of BMP signaling using an inhibitor of BMP receptor activity, dorsomorphin. Overall, these lines will serve as valuable tools to explore the mechanisms and regulation of BMP signal during embryogenesis, in tissue maintenance, and during disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Smad/metabolismo , Animales , Animales Modificados Genéticamente , Inmunohistoquímica , Microscopía Fluorescente , Elementos de Respuesta/genética , Elementos de Respuesta/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Pez Cebra
19.
Neural Dev ; 5: 12, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20412557

RESUMEN

BACKGROUND: Photoreceptors of the retina are highly compartmentalized cells that function as the primary sensory neurons for receiving and initiating transmission of visual information. Proper morphogenesis of photoreceptor neurons is essential for their normal function and survival. We have characterized a zebrafish mutation, cannonball, that completely disrupts photoreceptor morphogenesis. RESULTS: Analysis revealed a non-sense mutation in cytoplasmic dynein heavy chain 1 (dync1h1), a critical subunit in Dynein1, to underlie the cannonball phenotypes. Dynein1 is a large minus-end directed, microtubule motor protein complex that has been implicated in multiple, essential cellular processes. In photoreceptors, Dynein1 is thought to mediate post-Golgi vesicle trafficking, while Dynein2 is thought to be responsible for outer segment maintenance. Surprisingly, cannonball embryos survive until larval stages, owing to wild-type maternal protein stores. Retinal photoreceptor neurons, however, are significantly affected by loss of Dync1h1, as transmission electron microscopy and marker analyses demonstrated defects in organelle positioning and outer segment morphogenesis and suggested defects in post-Golgi vesicle trafficking. Furthermore, dosage-dependent antisense oligonucleotide knock-down of dync1h1 revealed outer segment abnormalities in the absence of overt inner segment polarity and trafficking defects. Consistent with a specific function of Dync1h1 within the outer segment, immunolocalization showed that this protein and other subunits of Dynein1 and Dynactin localized to the ciliary axoneme of the outer segment, in addition to their predicted inner segment localization. However, knock-down of Dynactin subunits suggested that this protein complex, which is known to augment many Dynein1 activities, is only essential for inner segment processes as outer segment morphogenesis was normal. CONCLUSIONS: Our results indicate that Dynein1 is required for multiple cellular processes in photoreceptor neurons, including organelle positioning, proper outer segment morphogenesis, and potentially post-Golgi vesicle trafficking. Titrated knock-down of dync1h1 indicated that outer segment morphogenesis was affected in photoreceptors that showed normal inner segments. These observations, combined with protein localization studies, suggest that Dynein1 may have direct and essential functions in photoreceptor outer segments, in addition to inner segment functions.


Asunto(s)
Dineínas Citoplasmáticas/genética , Dineínas/genética , Mutación/genética , Células Fotorreceptoras/metabolismo , Retina/crecimiento & desarrollo , Retina/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Animales , Axonema/metabolismo , Axonema/ultraestructura , Diferenciación Celular/genética , Dineínas Citoplasmáticas/antagonistas & inhibidores , Dineínas Citoplasmáticas/metabolismo , Dineínas/antagonistas & inhibidores , Regulación del Desarrollo de la Expresión Génica/genética , Microscopía Electrónica de Transmisión , Neurogénesis/fisiología , Oligonucleótidos Antisentido/farmacología , Células Fotorreceptoras/ultraestructura , Retina/ultraestructura , Vesículas Transportadoras/fisiología , Vesículas Transportadoras/ultraestructura
20.
J Urol ; 183(1): 133-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19913249

RESUMEN

PURPOSE: We report on outcomes of robotic assisted laparoscopic radical prostatectomy as salvage local therapy for radiation resistant prostate cancer. MATERIALS AND METHODS: We retrospectively reviewed the charts of all patients who underwent robotic assisted laparoscopic radical prostatectomy for biopsy proven prostate cancer after primary radiation treatment. Patient characteristics, intraoperative and perioperative data, and oncological and functional outcomes were assessed. RESULTS: A total of 18 patients were identified with a median followup of 18 months (range 4.5 to 40). Primary treatment was brachytherapy in 8 patients and external beam radiation in 8, while 2 underwent proton beam therapy. Median age at salvage robotic assisted laparoscopic radical prostatectomy was 67 years (range 53 to 76). Median preoperative prostate specific antigen was 6.8 ng/ml (range 1 to 28.9) and median time to surgery after primary treatment with radiation was 79 months (range 7 to 146). Median operative parameters for estimated blood loss, surgery length and hospital stay were 150 ml, 2.6 hours and 2 days, respectively. No patient required conversion to open surgery or a blood transfusion, or experienced a rectal injury. Perioperative complications occurred in 7 patients (39%) of which the most common was urine leak identified by postoperative cystogram. Five patients (28%) had a positive surgical margin. Although some patients had limited followup, 6 (33%) were continent and 67% were free of biochemical progression. CONCLUSIONS: Robotic assisted laparoscopic radical prostatectomy can be performed safely as salvage local therapy after failed radiation therapy. Outcomes are comparable to those of large series of open salvage prostatectomy.


Asunto(s)
Laparoscopía , Prostatectomía/métodos , Neoplasias de la Próstata/cirugía , Robótica , Anciano , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/radioterapia , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA