Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
BMC Pharmacol Toxicol ; 20(1): 67, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31730023

RESUMO

BACKGROUND: Neurofibromatosis 1 and 2, although involving two different tumour suppressor genes (neurofibromin and merlin, respectively), are both cancer predisposition syndromes that disproportionately affect cells of neural crest origin. New therapeutic approaches for both NF1 and NF2 are badly needed. In promising previous work we demonstrated that two non-steroidal analogues of 2-methoxy-oestradiol (2ME2), STX3451(2-(3-bromo-4,5-dimethoxybenzyl)-7-methoxy-6-sulfamoyloxy-1,2,3,4-tetrahydroisoquinoline), and STX2895 (7-Ethyl-6-sulfamoyloxy-2-(3,4,5-trimethoxybenzyl)-1,2,3,4-tetrahydroisoquinoline) reduced tumour cell growth and induced apoptosis in malignant and benign human Neurofibromatosis 1 (NF1) tumour cells. In earlier NF1 mechanism of action studies we found that in addition to their effects on non-classical hormone-sensitive pathways, STX agents acted on the actin- and myosin-cytoskeleton, as well as PI3Kinase and MTOR signaling pathways. Tumour growth in NF2 cells is affected by different inhibitors from those affecting NF1 growth pathways: specifically, NF2 cells are affected by merlin-downstream pathway inhibitors. Because Merlin, the affected tumour suppressor gene in NF2, is also known to be involved in stabilizing membrane-cytoskeletal complexes, as well as in cell proliferation, and apoptosis, we looked for potentially common mechanisms of action in the agents' effects on NF1 and NF2. We set out to determine whether STX agents could therefore also provide a prospective avenue for treatment of NF2. METHODS: STX3451 and STX2895 were tested in dose-dependent studies for their effects on growth parameters of malignant and benign NF2 human tumour cell lines in vitro. The mechanisms of action of STX3451 and STX2895 were also analysed. RESULTS: Although neither of the agents tested affected cell growth or apoptosis in the NF2 tumour cell lines tested through the same mechanisms by which they affect these parameters in NF1 tumour cell lines, both agents disrupted actin- and myosin-based cytoskeletal structures in NF2 cell lines, with subsequent effects on growth and cell death. CONCLUSIONS: Both STX3451 and STX2895 provide new approaches for inducing cell death and lowering tumour burden in NF2 as well as in NF1, which both have limited treatment options.


Assuntos
Antineoplásicos/farmacologia , Neurofibromatose 1/tratamento farmacológico , Neurofibromatose 2/tratamento farmacológico , Ácidos Sulfônicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Cicatrização/efeitos dos fármacos
2.
Exp Neurol ; 301(Pt B): 100-109, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28928022

RESUMO

Macrophage migration inhibitory factor (MIF) is a neurotrophic cytokine essential for inner ear hair cell (HC) development and statoacoustic ganglion (SAG) neurite outgrowth, and SAG survival in mouse, chick and zebrafish. Another neurotrophic cytokine, Monocyte chemoattractant protein 1 (MCP1) is known to synergize with MIF; but MCP1 alone is insufficient to support mouse/chick SAG neurite outgrowth or neuronal survival. Because of the relatively short time over which the zebrafish inner ear develops (~30hpf), the living zebrafish embryo is an ideal system to examine mif and mcp1 cytokine pathways and interactions. We used a novel technique: direct delivery of antisense oligonucleotide morpholinos (MOs) into the embryonic zebrafish otocyst to discover downstream effectors of mif as well as to clarify the relationship between mif and mcp1 in inner ear development. MOs for mif, mcp1 and the presumptive mif and mcp1 effector, c-Jun activation domain-binding protein-1 (jab1), were injected and then electroporated into the zebrafish otocyst 25-48hours post fertilization (hpf). We found that although mif is important at early stages (before 30hpf) for auditory macular HC development, jab1 is more critical for vestibular macular HC development before 30hpf. After 30hpf, mcp1 becomes important for HC development in both maculae.


Assuntos
Complexo do Signalossomo COP9/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Máculas Acústicas/embriologia , Máculas Acústicas/crescimento & desenvolvimento , Actinas/metabolismo , Animais , Axônios/efeitos dos fármacos , Complexo do Signalossomo COP9/genética , Quimiocina CCL2/metabolismo , Citocinas/biossíntese , Embrião não Mamífero , Fatores Inibidores da Migração de Macrófagos/genética , Oligonucleotídeos Antissenso/farmacologia , Oocistos/crescimento & desenvolvimento , Sáculo e Utrículo/embriologia , Sáculo e Utrículo/crescimento & desenvolvimento , Tubulina (Proteína)/metabolismo , Peixe-Zebra
3.
J Tissue Eng Regen Med ; 12(1): e379-e383, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27943657

RESUMO

New therapeutic approaches for repairing an injured or degenerating nervous system have accelerated the development of methods to generate populations of neurons derived from various stem cell sources efficiently. Many of these methods require the generation of neurospheres. Here a simple technique is described for creating an array of adherent mouse embryonic stem cell (mESC)-derived neurospheres using a conventional plastic culture dish and a patterning template. mESC-derived neurospheres are confined to circular (4-mm diameter), gel-coated regions within an array. The adherent neurosphere arrays require 3 days to prepare from an mESC source; they can be maintained in 15 µl drops of medium, and exhibit extensive neurite elaboration after 8 days of cultivation. Additionally, the potential of treating the adherent neurospheres in selected drops of an array is demonstrated with a variety of differentiation-inducing reagents and subsequently individually analysing such neurospheres for gene expression, protein levels and morphological development. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias Murinas/citologia , Neurônios/citologia , Esferoides Celulares/citologia , Animais , Adesão Celular , Camundongos , Neuritos/metabolismo , Neurônios/metabolismo , Esferoides Celulares/metabolismo
4.
Exp Neurol ; 301(Pt B): 92-99, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29080793

RESUMO

The earliest stages of neuronal and sensory cell development in vertebrate sensory organs depend on "inflammatory" immune system neurotrophic cytokines/chemokines. Although classical nerve growth factors, brain-derived neurotrophic factors and glial growth factors play critical roles at various stages, the earliest directive roles belong to immune system cytokines. In frogs, fishes, birds and mammals, macrophage migration inhibitory factor (MIF), monocyte chemoattractant protein 1 (MCP1) and RANTES, components of the otocyst-derived factor, are involved in sorting, morphogenesis, providing directional neuronal outgrowth cues as well as survival factors for both neurons and sensory cells. In this review we discuss their roles in the vertebrate inner ear.


Assuntos
Quimiocinas/metabolismo , Citocinas/metabolismo , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Sistema Imunitário/imunologia , Sistema Nervoso/imunologia , Animais , Humanos
5.
Dev Dyn ; 246(1): 7-27, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27761977

RESUMO

BACKGROUND: To send meaningful information to the brain, an inner ear cochlear implant (CI) must become closely coupled to as large and healthy a population of remaining spiral ganglion neurons (SGN) as possible. Inner ear gangliogenesis depends on macrophage migration inhibitory factor (MIF), a directionally attractant neurotrophic cytokine made by both Schwann and supporting cells (Bank et al., 2012). MIF-induced mouse embryonic stem cell (mESC)-derived "neurons" could potentially substitute for lost or damaged SGN. mESC-derived "Schwann cells" produce MIF, as do all Schwann cells (Huang et al., a; Roth et al., 2007; Roth et al., 2008) and could attract SGN to a "cell-coated" implant. RESULTS: Neuron- and Schwann cell-like cells were produced from a common population of mESCs in an ultra-slow-flow microfluidic device. As the populations interacted, "neurons" grew over the "Schwann cell" lawn, and early events in myelination were documented. Blocking MIF on the Schwann cell side greatly reduced directional neurite outgrowth. MIF-expressing "Schwann cells" were used to coat a CI: Mouse SGN and MIF-induced "neurons" grew directionally to the CI and to a wild-type but not MIF-knockout organ of Corti explant. CONCLUSIONS: Two novel stem cell-based approaches for treating the problem of sensorineural hearing loss are described. Developmental Dynamics 246:7-27, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular , Dispositivos Lab-On-A-Chip/normas , Células-Tronco Embrionárias Murinas/citologia , Neurônios/citologia , Células de Schwann/citologia , Animais , Implantes Cocleares/normas , Perda Auditiva/terapia , Oxirredutases Intramoleculares/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Camundongos , Bainha de Mielina/metabolismo , Gânglio Espiral da Cóclea
6.
Br J Cancer ; 113(8): 1158-67, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26461061

RESUMO

BACKGROUND: Both the number and size of tumours in NF1 patients increase in response to the rise in steroid hormones seen at puberty and during pregnancy. The size of tumours decreases after delivery, suggesting that hormone-targeting therapy might provide a viable new NF1 treatment approach. Our earlier studies demonstrated that human NF1 tumour cell lines either went through apoptosis or ceased growth in the presence of 2-methoxyoestradiol (2ME2), a naturally occurring anticancer metabolite of 17-ß estradiol. Previous reports of treatment with sulfamoylated steroidal and non-steroidal derivatives of 2ME2 showed promising reductions in tumour burden in hormone-responsive cancers other than NF1. Here we present the first studies indicating that 2ME2 derivatives could also provide an avenue for treating NF1, for which few treatment options are available. METHODS: STX3451, (2-(3-Bromo-4,5-dimethoxybenzyl)-7-methoxy-6-sulfamoyloxy-1,2,3,4-tetrahydroisoquinoline), a non-steroidal sulphamate analogue of 2ME2, was tested in dose-dependent studies of malignant and benign NF1 human tumour cell lines and cell lines with variable controlled neurofibromin expression. The mechanisms of action of STX3451 were also analysed. RESULTS: We found that STX3451-induced apoptosis in human malignant peripheral nerve sheath tumour (MPNST) cell lines, even in the presence of elevated oestrogen and progesterone. It inhibits both PI3 kinase and mTOR signalling pathways. It disrupts actin- and microtubule-based cytoskeletal structures in cell lines derived from human MPNSTs and in cells derived from benign plexiform neurofibromas. STX3451 selectively kills MPNST-derived cells, but also halts growth of other tumour-derived NF1 cell lines. CONCLUSION: STX3451 provides a new approach for inducing cell death and lowering tumour burden in NF1 and other hormone-responsive cancers with limited treatment options.


Assuntos
Antineoplásicos/farmacologia , Estradiol/análogos & derivados , Estradiol/metabolismo , Neurofibroma Plexiforme/tratamento farmacológico , Neurofibromatose 1/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , 2-Metoxiestradiol , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Estradiol/farmacologia , Estrogênios/metabolismo , Humanos , Neurofibroma Plexiforme/metabolismo , Neurofibromatose 1/metabolismo , Neurofibromina 1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
7.
Expert Rev Mol Diagn ; 15(9): 1171-86, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26289603

RESUMO

Methods to detect immunolabeled molecules at increasingly higher resolutions, even when present at low levels, are revolutionizing immunohistochemistry (IHC). These technologies can be valuable for the management and examination of rare patient tissue specimens, and for improved accuracy of early disease detection. The purpose of this article is to highlight recent multiplexing methods that are candidates for more prevalent use in clinical research and potential translation to the clinic. Multiplex IHC methods, which permit identification of at least 3 and up to 30 discrete antigens, have been divided into whole-section staining and spatially-patterned staining categories. Associated signal enhancement technologies that can enhance performance and throughput of multiplex IHC assays are also discussed. Each multiplex IHC technique, detailed herein, is associated with several advantages as well as tradeoffs that must be taken into consideration for proper evaluation and use of the methods.


Assuntos
Imuno-Histoquímica/métodos , Humanos , Análise em Microsséries/métodos , Microfluídica/métodos
8.
Dev Dyn ; 243(11): 1487-98, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25178196

RESUMO

BACKGROUND: Murine Zic genes (Zic1-5) are expressed in the dorsal hindbrain and in periotic mesenchyme (POM) adjacent to the developing inner ear. Zic genes are involved in developmental signaling pathways in many organ systems, including the ear, although their exact roles haven't been fully elucidated. This report examines the role of Zic1, Zic2, and Zic4 during inner ear development in mouse mutants in which these Zic genes are affected. RESULTS: Zic1/Zic4 double mutants don't exhibit any apparent defects in inner ear morphology. By contrast, inner ears from Zic2(kd/kd) and Zic2(Ku/Ku) mutants have severe but variable morphological defects in endolymphatic duct/sac and semicircular canal formation and in cochlear extension in the inner ear. Analysis of otocyst patterning in the Zic2(Ku/Ku) mutants by in situ hybridization showed changes in the expression patterns of Gbx2 and Pax2. CONCLUSIONS: The experiments provide the first genetic evidence that the Zic genes are required for morphogenesis of the inner ear. Zic2 loss-of-function doesn't prevent initial otocyst patterning but leads to molecular abnormalities concomitant with morphogenesis of the endolymphatic duct. Functional hearing deficits often accompany inner ear dysmorphologies, making Zic2 a novel candidate gene for ongoing efforts to identify the genetic basis of human hearing loss.


Assuntos
Orelha Interna/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/genética , Morfogênese/fisiologia , Fenótipo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Animais , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Camundongos , Mutação/genética , Fatores de Transcrição/metabolismo
9.
Sci Rep ; 3: 1921, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23719665

RESUMO

Alzheimer's disease is accompanied by progressive, time-dependent changes of three moieties of amyloid beta. In vitro models therefore should provide same conditions for more physiologic studies. Here we observed changes in the number of fibrils over time and studied the correlation between amyloid beta moieties and neurotoxicity. Although the number of fibrils increased dramatically, the change in neurotoxicity with time was small, suggesting that fibrils make little contribution to neurotoxicity. To study the neurotoxicity of diffusible moieties by regulating microenvironments, we created a bio-mimetic microfluidic system generating spatial gradients of diffusible oligomeric assemblies and assessed their effects on cultured neurons. We found amyloid beta exposure produced an atrophy effect and observed neurite extension during the differentiation of neural progenitor cells increased when cells were cultured with continuous flow. The results demonstrate the potential neurotoxicity of oligomeric assemblies and establish a prospective microfluidic platform for studying the neurotoxicity of amyloid beta.


Assuntos
Peptídeos beta-Amiloides/química , Amiloide/química , Microfluídica/métodos , Peptídeos beta-Amiloides/toxicidade , Animais , Apoptose/efeitos dos fármacos , Biomimética/instrumentação , Biomimética/métodos , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Microfluídica/instrumentação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Sinapsinas/metabolismo
10.
Dev Dyn ; 242(7): 897-908, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23606270

RESUMO

BACKGROUND: Inner ear development involves signaling from surrounding tissues, including the adjacent hindbrain, periotic mesenchyme, and notochord. These signals include SHH, FGFs, BMPs, and WNTs from the hindbrain and SHH from the notochord. Zic genes, which are expressed in the dorsal neural tube and act during neural development, have been implicated as effectors of these pathways. This report examines whether Zic genes' involvement in inner ear development is a tenable hypothesis based on their expression patterns. RESULTS: In the developing inner ear of both the chick and mouse, all of the Zic genes were expressed in the dorsal neural tube and variably in the periotic mesenchyme, but expression of the Zic genes in the otic epithelium was not found. The onset of expression differed among the Zic genes; within any given region surrounding the otic epithelium, multiple Zic genes were expressed in the same place at the same time. CONCLUSIONS: Zic gene expression in the region of the developing inner ear is similar between mouse and chick. Zic expression domains overlap with sites of WNT and SHH signaling during otocyst patterning, suggesting a role for Zic genes in modulating signaling from these pathways.


Assuntos
Orelha Interna/embriologia , Orelha Interna/metabolismo , Vertebrados/embriologia , Vertebrados/metabolismo , Animais , Embrião de Galinha , Galinhas , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Development ; 139(24): 4666-74, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23172918

RESUMO

This study is the first to demonstrate that macrophage migration inhibitory factor (MIF), an immune system 'inflammatory' cytokine that is released by the developing otocyst, plays a role in regulating early innervation of the mouse and chick inner ear. We demonstrate that MIF is a major bioactive component of the previously uncharacterized otocyst-derived factor, which directs initial neurite outgrowth from the statoacoustic ganglion (SAG) to the developing inner ear. Recombinant MIF acts as a neurotrophin in promoting both SAG directional neurite outgrowth and neuronal survival and is expressed in both the developing and mature inner ear of chick and mouse. A MIF receptor, CD74, is found on both embryonic SAG neurons and adult mouse spiral ganglion neurons. Mif knockout mice are hearing impaired and demonstrate altered innervation to the organ of Corti, as well as fewer sensory hair cells. Furthermore, mouse embryonic stem cells become neuron-like when exposed to picomolar levels of MIF, suggesting the general importance of this cytokine in neural development.


Assuntos
Orelha Interna/embriologia , Oxirredutases Intramoleculares/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Fatores de Crescimento Neural/fisiologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Orelha Interna/efeitos dos fármacos , Orelha Interna/crescimento & desenvolvimento , Orelha Interna/metabolismo , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Oxirredutases Intramoleculares/farmacologia , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/metabolismo , Fatores Inibidores da Migração de Macrófagos/farmacologia , Camundongos , Camundongos Knockout , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Órgão Espiral/embriologia , Órgão Espiral/crescimento & desenvolvimento , Órgão Espiral/metabolismo , Gânglio Espiral da Cóclea/embriologia , Gânglio Espiral da Cóclea/crescimento & desenvolvimento , Gânglio Espiral da Cóclea/metabolismo
12.
Dev Biol ; 363(1): 84-94, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22210003

RESUMO

Macrophage migration inhibitory factor (MIF) plays versatile roles in the immune system. MIF is also widely expressed during embryonic development, particularly in the nervous system, although its roles in neural development are only beginning to be understood. Evidence from frogs, mice and zebrafish suggests that MIF has a major role as a neurotrophin in the early development of sensory systems, including the auditory system. Here we show that the zebrafish mif pathway is required for both sensory hair cell (HC) and sensory neuronal cell survival in the ear, for HC differentiation, semicircular canal formation, statoacoustic ganglion (SAG) development, and lateral line HC differentiation. This is consistent with our findings that MIF is expressed in the developing mammalian and avian auditory systems and promotes mouse and chick SAG neurite outgrowth and neuronal survival, demonstrating key instructional roles for MIF in vertebrate otic development.


Assuntos
Orelha Interna/metabolismo , Fatores Inibidores da Migração de Macrófagos/genética , Fatores de Crescimento Neural/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Diferenciação Celular/genética , Orelha Interna/embriologia , Embrião não Mamífero/citologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Gânglios Sensitivos/embriologia , Gânglios Sensitivos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células Ciliadas Auditivas/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Pirimidinas/farmacologia , Receptores Imunológicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais Semicirculares/embriologia , Canais Semicirculares/metabolismo , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Fatores de Tempo , Peixe-Zebra/embriologia
13.
J Vis Exp ; (47)2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21248702

RESUMO

In recent years, electroporation has become a popular technique for in vivo transfection of DNA, RNA, and morpholinos into various tissues, including the eye, brain, and somites of zebrafish. The advantage of electroporation over other methods of genetic manipulation is that specific tissues can be targeted, both spatially and temporally, for the introduction of macromolecules by the application of electrical current. Here we describe the use of electroporation for transfecting mif and mif-like morpholinos into the tissues of the developing inner ear of the zebrafish. In past studies, mif morpholino injected into embryos at the 1- to 8-cell stage resulted in widespread morphological changes in the nervous system and eye, as well as the ear. By targeting the tissues of the inner ear at later stages in development, we can determine the primary effects of MIF in the developing inner ear, as opposed to secondary effects that may result from the influence of other tissues. By using phalloidin and acetylated tubulin staining to study the morphology of neurons, neuronal processes, and hair cells associated with the posterior macula, we were able to assess the efficacy of electroporation as a method for targeted transfection in the zebrafish inner ear. The otic vesicles of 24hpf embryos were injected with morpholinos and electroporated and were then compared to embryos that had received no treatment or had been only injected or electroporated. Embryos that were injected and electroporated showed a decrease in hair cell numbers, decreased innervation by the statoacoustic ganglion (SAG) and fewer SAG neurons compared with control groups. Our results showed that direct delivery of morpholinos into otocysts at later stages avoids the non-specific nervous system and neural crest effects of morpholinos delivered at the 1-8 cell stage. It also allows examination of effects that are directed to the inner ear and not secondary effects on the ear from primary effects on the brain, neural crest or periotic mesenchyme.


Assuntos
Eletroporação/métodos , Fatores Inibidores da Migração de Macrófagos/genética , Morfolinas/administração & dosagem , Oligonucleotídeos Antissenso/administração & dosagem , Transfecção/métodos , Animais , Orelha Interna/citologia , Peixe-Zebra
14.
Lab Chip ; 10(21): 2959-64, 2010 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-20835429

RESUMO

Generation of stable soluble-factor gradients in microfluidic devices enables studies of various cellular events such as chemotaxis and differentiation. However, many gradient devices directly expose cells to constant fluid flow and that can induce undesired responses from cells due to shear stress and/or wash out of cell-secreted molecules. Although there have been devices with flow-free gradients, they typically generate only a single condition and/or have a decaying gradient profile that does not accommodate long-term experiments. Here we describe a microdevice that generates several chemical gradient conditions on a single platform in flow-free microchambers which facilitates steady-state gradient profiles. The device contains embedded normally-closed valves that enable fast and uniform seeding of cells to all microchambers simultaneously. A network of microchannels distributes desired solutions from easy-access open reservoirs to a single output port, enabling a simple setup for inducing flow in the device. Embedded porous filters, sandwiched between the microchannel networks and cell microchambers, enable diffusion of biomolecules but inhibit any bulk flow over the cells.


Assuntos
Microfluídica/instrumentação , Animais , Células-Tronco Embrionárias/citologia , Camundongos
15.
Dev Dyn ; 238(8): 1909-22, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19582870

RESUMO

Cadherin-11/Cdh11 is expressed through early development and strongly during inner ear development (otic placode and vesicle). Here we show that antisense knockdown of Cdh11 during early zebrafish development interferes with otolith formation. Immunofluorescence labeling showed that Cdh11 expression was concentrated on and within the otolith. Cdh11 was faintly detected at the lateral surface (sites of cell-cell contact) of otic epithelial cells and in the cytoplasm. Strongly labeled Cdh11 containing puncta were detected within the otolymph (the fluid within the otic vesicle) and associated with the otolith surface. BODIPY-ceramine-labeled vesicular structures detected in the otolymph were larger and more numerous in Cdh11 knockdown embryos. We present evidence supporting a working model that vesicular structures containing Cdh11 (perhaps containing biomineralization components) are exported from the otic epithelium into the otolymph, adhere to one another and to the surface of the growing otolith, facilitating otolith growth.


Assuntos
Caderinas/metabolismo , Membrana dos Otólitos/embriologia , Membrana dos Otólitos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Caderinas/deficiência , Caderinas/genética , Espaço Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Modelos Biológicos , Dados de Sequência Molecular , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rombencéfalo/anormalidades , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
16.
Zebrafish ; 6(2): 201-13, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19292670

RESUMO

The zebrafish is a valuable model for teaching developmental, molecular, and cell biology; aquatic sciences; comparative anatomy; physiology; and genetics. Here we demonstrate that zebrafish provide an excellent model system to teach engineering principles. A seven-member undergraduate team in a biomedical engineering class designed, built, and tested a zebrafish microfluidic bioreactor applying microfluidics, an emerging engineering technology, to study zebrafish development. During the semester, students learned engineering and biology experimental design, chip microfabrication, mathematical modeling, zebrafish husbandry, principles of developmental biology, fluid dynamics, microscopy, and basic molecular biology theory and techniques. The team worked to maximize each person's contribution and presented weekly written and oral reports. Two postdoctoral fellows, a graduate student, and three faculty instructors coordinated and directed the team in an optimal blending of engineering, molecular, and developmental biology skill sets. The students presented two posters, including one at the Zebrafish meetings in Madison, Wisconsin (June 2008).


Assuntos
Engenharia Biomédica/educação , Técnicas Analíticas Microfluídicas , Peixe-Zebra/embriologia , Animais , Engenharia Biomédica/métodos , Universidades
17.
Am J Med Genet A ; 146A(12): 1624-33, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18481270

RESUMO

The Neurofibromatosis Type 1 (NF1) gene functions as a tumor suppressor gene. Loss of its protein, neurofibromin, in the autosomal dominant disorder NF1 is associated with peripheral nervous system tumors, particularly neurofibromas, benign lesions in which the major cell type is the Schwann Cell (SC). Benign and malignant human tumors found in NF1 patients are heterogeneous with respect to their cellular composition. The number and size of neurofibromas in NF1 patients has been shown to increase during pregnancy, with, in some cases, post-partum regression, which suggests hormonal involvement in this increase. However, in this review, we consider evidence from the literature that both direct hormonal influence on tumor growth and on angiogenesis may contribute to these effects.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Neurofibromatose 1/genética , Neurofibromatose 1/metabolismo , Neurofibromina 1/genética , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Apoptose/genética , Vasos Sanguíneos/ultraestrutura , Feminino , Humanos , Hipertensão Induzida pela Gravidez/genética , Hipertensão Induzida pela Gravidez/metabolismo , Fenótipo , Gravidez
18.
Dev Dyn ; 237(4): 941-52, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18330929

RESUMO

The inner ear is a complex organ containing sensory tissue, including hair cells, the development of which is not well understood. Our long-term goal is to discover genes critical for the correct formation and function of the inner ear and its sensory tissue. A novel gene, transmembrane inner ear (Tmie), was found to cause hearing-related disorders when defective in mice and humans. A homologous tmie gene in zebrafish was cloned and its expression characterized between 24 and 51 hours post-fertilization. Embryos injected with morpholinos (MO) directed against tmie exhibited circling swimming behavior (approximately 37%), phenocopying mice with Tmie mutations; semicircular canal formation was disrupted, hair cell numbers were reduced, and maturation of electrically active lateral line neuromasts was delayed. As in the mouse, tmie appears to be required for inner ear development and function in the zebrafish and for hair cell maturation in the vestibular and lateral line systems as well.


Assuntos
Orelha Interna/embriologia , Orelha Interna/fisiologia , Sistema da Linha Lateral/embriologia , Sistema da Linha Lateral/fisiologia , Proteínas de Membrana/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra , Sequência de Aminoácidos , Animais , Comportamento Animal/fisiologia , Orelha Interna/anatomia & histologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Sistema da Linha Lateral/anatomia & histologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Morfogênese , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Alinhamento de Sequência , Natação/fisiologia , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/metabolismo
19.
Dev Dyn ; 237(2): 513-24, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18213578

RESUMO

Loss of neurofibromin, the protein product of the tumor suppressor gene neurofibromatosis type 1 (NF1), is associated with neurofibromas, composed largely of Schwann cells. The number and size of neurofibromas in NF1 patients have been shown to increase during pregnancy. A mouse embryonic stem cell (mESC) model was used, in which mESCs with varying levels of neurofibromin were differentiated into Schwann-like cells. NF1 cell lines derived from a malignant and a benign human tumor were used to study proliferation in response to hormones. Estrogen and androgen receptors were not expressed or expressed at very low levels in the NF1+/+ cells, at low levels in NF1+/-cells, and robust levels in NF1-/-cells. A 17beta-estradiol (E2) metabolite, 2-methoxy estradiol (2ME2) is cytotoxic to the NF1-/- malignant tumor cell line, and inhibits proliferation in the other cell lines. 2ME2 or its derivatives could provide new treatment avenues for NF1 hormone-sensitive tumors at times of greatest hormonal influence.


Assuntos
Células-Tronco Embrionárias/fisiologia , Estradiol/análogos & derivados , Neurofibroma/fisiopatologia , Neurofibromina 1/metabolismo , Células de Schwann/efeitos dos fármacos , Células de Schwann/fisiologia , 2-Metoxiestradiol , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Primers do DNA/genética , Células-Tronco Embrionárias/citologia , Estradiol/toxicidade , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Neurofibroma/metabolismo , Neurofibromina 1/genética , Receptores de Estradiol/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Schwann/citologia
20.
Lab Chip ; 7(6): 770-6, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17538720

RESUMO

The formation of spherical aggregates of cells called embryoid bodies (EBs) is an indispensable step in many protocols in which embryonic stem (ES) cells are differentiated to other cell types. Appropriate morphology and embryo size are critical for the sequential developmental stages of naturally conceived embryos. Likewise, regulating the size of EBs and the timing of their formation is crucial for controlling the differentiation of ES cells within the EB. Existing methods of formation of EBs, however, are tedious or provide heterogeneously-sized EBs. Here we describe a microfluidic system for straightforward synchronized formation of uniform-sized EBs, the size of which can be controlled by changing the cross-sectional size of microchannels in the microfluidic device. The device consists of two microchannels separated by a semi-porous polycarbonate membrane treated to be resistant to cell adhesion. ES cells introduced into the upper channel self-aggregate to form uniformly-sized EBs. The semi-porous membrane also allows subsequent treatment of the non-attached EBs with different reagents from the lower channel without the need for wash out because of the compartmentalization afforded by the membrane. This method provides a simple yet robust means to control the formation of EBs and the subsequent differentiation of ES cells in a format compatible for ES cell processing on a chip.


Assuntos
Embrião de Mamíferos/citologia , Técnicas Analíticas Microfluídicas , Engenharia Tecidual , Animais , Adesão Celular/fisiologia , Agregação Celular/fisiologia , Células Cultivadas , Embrião de Mamíferos/fisiologia , Embrião de Mamíferos/ultraestrutura , Camundongos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA