Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(46): e2302089120, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37931105

RESUMO

Ongoing cell therapy trials have demonstrated the need for precision control of donor cell behavior within the recipient tissue. We present a methodology to guide stem cell-derived and endogenously regenerated neurons by engineering the microenvironment. Being an "approachable part of the brain," the eye provides a unique opportunity to study neuron fate and function within the central nervous system. Here, we focused on retinal ganglion cells (RGCs)-the neurons in the retina are irreversibly lost in glaucoma and other optic neuropathies but can potentially be replaced through transplantation or reprogramming. One of the significant barriers to successful RGC integration into the existing mature retinal circuitry is cell migration toward their natural position in the retina. Our in silico analysis of the single-cell transcriptome of the developing human retina identified six receptor-ligand candidates, which were tested in functional in vitro assays for their ability to guide human stem cell-derived RGCs. We used our lead molecule, SDF1, to engineer an artificial gradient in the retina, which led to a 2.7-fold increase in donor RGC migration into the ganglion cell layer (GCL) and a 3.3-fold increase in the displacement of newborn RGCs out of the inner nuclear layer. Only donor RGCs that migrated into the GCL were found to express mature RGC markers, indicating the importance of proper structure integration. Together, these results describe an "in silico-in vitro-in vivo" framework for identifying, selecting, and applying soluble ligands to control donor cell function after transplantation.


Assuntos
Retina , Células Ganglionares da Retina , Recém-Nascido , Humanos , Células-Tronco , Neurogênese , Movimento Celular
2.
Ophthalmol Sci ; 3(4): 100390, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38025164

RESUMO

Purpose: The Retinal Ganglion Cell (RGC) Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) consortium was founded in 2021 to help address the numerous scientific and clinical obstacles that impede development of vision-restorative treatments for patients with optic neuropathies. The goals of the RReSTORe consortium are: (1) to define and prioritize the most critical challenges and questions related to RGC regeneration; (2) to brainstorm innovative tools and experimental approaches to meet these challenges; and (3) to foster opportunities for collaborative scientific research among diverse investigators. Design and Participants: The RReSTORe consortium currently includes > 220 members spanning all career stages worldwide and is directed by an organizing committee comprised of 15 leading scientists and physician-scientists of diverse backgrounds. Methods: Herein, we describe the structure and organization of the RReSTORe consortium, its activities to date, and the perceived impact that the consortium has had on the field based on a survey of participants. Results: In addition to helping propel the field of regenerative medicine as applied to optic neuropathies, the RReSTORe consortium serves as a framework for developing large collaborative groups aimed at tackling audacious goals that may be expanded beyond ophthalmology and vision science. Conclusions: The development of innovative interventions capable of restoring vision for patients suffering from optic neuropathy would be transformative for the ophthalmology field, and may set the stage for functional restoration in other central nervous system disorders. By coordinating large-scale, international collaborations among scientists with diverse and complementary expertise, we are confident that the RReSTORe consortium will help to accelerate the field toward clinical translation. Financial Disclosures: Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.

3.
Mol Neurodegener ; 18(1): 64, 2023 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-37735444

RESUMO

Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.


Assuntos
Doenças do Nervo Óptico , Células Ganglionares da Retina , Animais , Humanos , Retina , Encéfalo , Diferenciação Celular , Mamíferos
4.
Transl Vis Sci Technol ; 6(3): 4, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28516002

RESUMO

PURPOSE: Previous studies have demonstrated the ability of retinal cells derived from human embryonic stem cells (hESCs) to survive, integrate into the host retina, and mediate light responses in murine mouse models. Our aim is to determine whether these cells can also survive and integrate into the retina of a nonhuman primate, Saimiri sciureus, following transplantation into the subretinal space. METHODS: hESCs were differentiated toward retinal neuronal fates using our previously published technique and cultured for 60 to 70 days. Differentiated cells were further treated with 20 µM N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) for a period of 5 days immediately prior to subretinal transplantation. Differentiated cells were labeled with a lentivirus expressing GFP. One million cells (10,000 cells/µL) were injected into the submacular space into a squirrel monkey eye, using an ab externo technique. RESULTS: RetCam imaging demonstrated the presence and survival of human donor cells 3 months after transplantation in the S. sciureus eye. Injected cells consolidated in the temporal macula. GFP+ axonal projections were observed to emanate from the central consolidation of cells at 1 month, with some projecting into the optic nerve by 3 months after transplantation. CONCLUSIONS: Human ES cell-derived retinal neurons injected into the submacular space of a squirrel monkey survive at least 3 months postinjection without immunosuppression. Some donor cells appeared to integrate into the host inner retina, and numerous donor axonal projections were noted throughout, with some projecting into the optic nerve. TRANSLATIONAL RELEVANCE: These data illustrate the feasibility of hESC-derived retinal cell replacement in the nonhuman primate eye.

5.
Stem Cells ; 33(9): 2674-85, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26013465

RESUMO

During early patterning of the neural plate, a single region of the embryonic forebrain, the eye field, becomes competent for eye development. The hallmark of eye field specification is the expression of the eye field transcription factors (EFTFs). Experiments in fish, amphibians, birds, and mammals have demonstrated largely conserved roles for the EFTFs. Although some of the key signaling events that direct the synchronized expression of these factors to the eye field have been elucidated in fish and frogs, it has been more difficult to study these mechanisms in mammalian embryos. In this study, we have used two different methods for directed differentiation of mouse embryonic stem cells (mESCs) to generate eye field cells and retina in vitro to test for a role of the PDZ domain-containing protein GIPC1 in the specification of the mammalian eye primordia. We find that the overexpression of a dominant-negative form of GIPC1 (dnGIPC1), as well as the downregulation of endogenous GIPC1, is sufficient to inhibit the development of eye field cells from mESCs. GIPC1 interacts directly with IGFR and participates in Akt1 activation, and pharmacological inhibition of Akt1 phosphorylation mimics the dnGIPC1 phenotype. Our data, together with previous studies in Xenopus, support the hypothesis that the GIPC1-PI3K-Akt1 pathway plays a key role in eye field specification in vertebrates.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas do Olho/biossíntese , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/biossíntese , Retina/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Células HEK293 , Humanos , Camundongos , Retina/citologia , Xenopus laevis
7.
Stem Cells ; 31(9): 1737-48, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23765801

RESUMO

Adult stem cells reside in hypoxic niches, and embryonic stem cells (ESCs) are derived from a low oxygen environment. However, it is not clear whether hypoxia is critical for stem cell fate since for example human ESCs (hESCs) are able to self-renew in atmospheric oxygen concentrations as well. We now show that hypoxia can govern cell fate decisions since hypoxia alone can revert hESC- or iPSC-derived differentiated cells back to a stem cell-like state, as evidenced by re-activation of an Oct4-promoter reporter. Hypoxia-induced "de-differentiated" cells also mimic hESCs in their morphology, long-term self-renewal capacity, genome-wide mRNA and miRNA profiles, Oct4 promoter methylation state, cell surface markers TRA1-60 and SSEA4 expression, and capacity to form teratomas. These data demonstrate that hypoxia can influence cell fate decisions and could elucidate hypoxic niche function.


Assuntos
Linhagem da Célula , Células-Tronco Pluripotentes/citologia , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Desdiferenciação Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Histona Desacetilases/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Modelos Biológicos , Fator 3 de Transcrição de Octâmero/metabolismo , Oxigênio/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo
8.
Glia ; 60(10): 1579-89, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22777914

RESUMO

Müller glia are normally mitotically quiescent cells, but in certain pathological states they can re-enter the mitotic cell cycle. While several cell cycle regulators have been shown to be important in this process, a role for the tumor suppressor, p53, has not been demonstrated. Here, we investigated a role for p53 in limiting the ability of Müller glia to proliferate in the mature mouse retina. Our data demonstrate that Müller glia undergo a developmental restriction in their potential to proliferate. Retinal explants or dissociated cultures treated with EGF become mitotically quiescent by the end of the second postnatal week. In contrast, Müller glia from adult trp53-/+ or trp53-/- mice displayed a greater ability to proliferate in response to EGF stimulation in vitro. The enhanced proliferative ability of trp53 deficient mice correlates with a decreased expression of the mitotic inhibitor Cdkn1a/p21(cip) and an increase in c-myc, a transcription factor that promotes cell cycle progression. These data show that p53 plays an essential role in limiting the potential of Müller glia to re-enter the mitotic cycle as the retina matures during postnatal development.


Assuntos
Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento/genética , Neuroglia/fisiologia , Retina/citologia , Retina/crescimento & desenvolvimento , Proteína Supressora de Tumor p53/metabolismo , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Proteína 1 Inibidora de Diferenciação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/efeitos dos fármacos , Técnicas de Cultura de Órgãos , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Fatores de Tempo , Proteína Supressora de Tumor p53/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
9.
Methods Mol Biol ; 884: 229-46, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22688710

RESUMO

Over the last few years, numerous studies have introduced strategies for the generation of neuronal populations from embryonic stem cells. These techniques are valuable both in the study of early neurogenesis and in the generation of an unlimited source of donor cells for replacement therapies. We have developed a protocol to direct mouse and human embryonic stem cells to retinal fates by using the current model of eye specification. Our method is a multistep protocol in which the cultures are treated with IGF1 and a combination of BMP and Wnt inhibitors to promote the expression of key retinal progenitor genes, as assayed by RT-PCR and immunofluorescence microscopy. The retinal progenitor population spontaneously undergoes differentiation towards various types of retinal neurons, including photoreceptors.


Assuntos
Células-Tronco Embrionárias/citologia , Retina/citologia , Transplante de Células-Tronco , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Imuno-Histoquímica , Camundongos , Microinjeções , Reação em Cadeia da Polimerase em Tempo Real , Retina/metabolismo
10.
PLoS One ; 5(1): e8763, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20098701

RESUMO

BACKGROUND: Inherited and acquired retinal degenerations are frequent causes of visual impairment and photoreceptor cell replacement therapy may restore visual function to these individuals. To provide a source of new retinal neurons for cell based therapies, we developed methods to derive retinal progenitors from human ES cells. METHODOLOGY/PHYSICAL FINDINGS: In this report we have used a similar method to direct induced pluripotent stem cells (iPS) from human fibroblasts to a retinal progenitor fate, competent to generate photoreceptors. We also found we could purify the photoreceptors derived from the iPS cells using fluorescence activated cell sorting (FACS) after labeling photoreceptors with a lentivirus driving GFP from the IRBP cis-regulatory sequences. Moreover, we found that when we transplanted the FACS purified iPSC derived photoreceptors, they were able to integrate into a normal mouse retina and express photoreceptor markers. CONCLUSIONS: This report provides evidence that enriched populations of human photoreceptors can be derived from iPS cells.


Assuntos
Células Fotorreceptoras de Vertebrados/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transplante de Células-Tronco , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase
11.
Prog Brain Res ; 175: 23-31, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19660646

RESUMO

The retina, like most other regions of the central nervous system, is subject to degeneration from both genetic and acquired causes. Once the photoreceptors or inner retinal neurons have degenerated, they are not spontaneously replaced in mammals. In this review, we provide an overview of retinal development and regeneration with emphasis on endogenous repair and replacement seen in lower vertebrates and recent work on induced mammalian retinal regeneration from Müller glia. Additionally, recent studies demonstrating the potential for cellular replacement using postmitotic photoreceptors and embryonic stem cells are also reviewed.


Assuntos
Regeneração Nervosa/fisiologia , Retina/fisiologia , Degeneração Retiniana/cirurgia , Transplante de Células-Tronco/métodos , Animais , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Humanos
12.
Cell Stem Cell ; 4(1): 73-9, 2009 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-19128794

RESUMO

Some of the most common causes of blindness involve the degeneration of photoreceptors in the neural retina; photoreceptor replacement therapy might restore some vision in these individuals. Embryonic stem cells (ESCs) could, in principle, provide a source of photoreceptors to repair the retina. We have previously shown that retinal progenitors can be efficiently derived from human ESCs. We now show that retinal cells derived from human ESCs will migrate into mouse retinas following intraocular injection, settle into the appropriate layers, and express markers for differentiated cells, including both rod and cone photoreceptor cells. After transplantation of the cells into the subretinal space of adult Crx(-/-) mice (a model of Leber's Congenital Amaurosis), the hESC-derived retinal cells differentiate into functional photoreceptors and restore light responses to the animals. These results demonstrate that hESCs can, in principle, be used for photoreceptor replacement therapies.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Células Fotorreceptoras de Vertebrados/transplante , Transativadores/deficiência , Visão Ocular/fisiologia , Animais , Animais Recém-Nascidos , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Células Fotorreceptoras de Vertebrados/citologia , Retina/citologia , Retina/transplante , Transativadores/metabolismo
13.
Proc Natl Acad Sci U S A ; 105(49): 19508-13, 2008 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-19033471

RESUMO

Müller glia can serve as a source of new neurons after retinal damage in both fish and birds. Investigations of regeneration in the mammalian retina in vitro have provided some evidence that Müller glia can proliferate after retinal damage and generate new rods; however, the evidence that this occurs in vivo is not conclusive. We have investigated whether Müller glia have the potential to generate neurons in the mouse retina in vivo by eliminating ganglion and amacrine cells with intraocular NMDA injections and stimulating Müller glial to re-enter the mitotic cycle by treatment with specific growth factors. The proliferating Müller glia dedifferentiate and a subset of these cells differentiated into amacrine cells, as defined by the expression of amacrine cell-specific markers Calretinin, NeuN, Prox1, and GAD67-GFP. These results show for the first time that the mammalian retina has the potential to regenerate inner retinal neurons in vivo.


Assuntos
Regeneração Nervosa/fisiologia , Neuroglia/citologia , Neurônios/citologia , Retina/citologia , Retina/fisiologia , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Biomarcadores/metabolismo , Calbindina 2 , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Linhagem da Célula/fisiologia , Proteínas de Ligação a DNA , Denervação , Agonistas de Aminoácidos Excitatórios/toxicidade , Glutamato Descarboxilase/metabolismo , Proteínas de Fluorescência Verde , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Transgênicos , N-Metilaspartato/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Proteínas Supressoras de Tumor/metabolismo
14.
Stem Cells ; 25(8): 2033-43, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17525239

RESUMO

Growing evidence suggests that glial cells may have a role as neural precursors in the adult central nervous system. Although it has been shown that Müller cells exhibit progenitor characteristics in the postnatal chick and rat retinae, their progenitor-like role in developed human retina is unknown. We first reported the Müller glial characteristics of the spontaneously immortalized human cell line MIO-M1, but recently we have derived similar cell lines from the neural retina of several adult eye donors. Since immortalization is one of the main properties of stem cells, we investigated whether these cells expressed stem cell markers. Cells were grown as adherent monolayers, responded to epidermal growth factor, and could be expanded indefinitely without growth factors under normal culture conditions. They could be frozen and thawed without losing their characteristics. In the presence of extracellular matrix and fibroblast growth factor-2 or retinoic acid, they acquired neural morphology, formed neurospheres, and expressed neural stem cell markers including betaIII tubulin, Sox2, Pax6, Chx10, and Notch 1. They also expressed markers of postmitotic retinal neurons, including peripherin, recoverin, calretinin, S-opsin, and Brn3. When grafted into the subretinal space of dystrophic Royal College of Surgeons rats or neonatal Lister hooded rats, immortalized cells migrated into the retina, where they expressed various markers of retinal neurons. These observations indicate that adult human neural retina harbors a population of cells that express both Müller glial and stem cell markers and suggest that these cells may have potential use for cell-based therapies to restore retinal function. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Neuroglia/citologia , Neurônios/citologia , Retina/citologia , Células-Tronco/citologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Animais Recém-Nascidos , Biomarcadores/análise , Diferenciação Celular , Linhagem Celular Transformada , Células Cultivadas , Criança , Pré-Escolar , Humanos , Lactente , Pessoa de Meia-Idade , Ratos , Retina/transplante , Transplante Heterólogo
16.
Glia ; 54(2): 94-104, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16710850

RESUMO

Epidermal growth factor (EGF) is known to promote proliferation of both retinal progenitors and Muller glia in vitro, but several questions remain concerning an in vivo role for this factor. In this study, we investigated whether the EGF receptor (EGFR) is necessary for the maintenance of normal levels of progenitor and Muller glial proliferation in vivo. Here, we show that (1) mice with homozygous deletion of the Egfr gene have reduced proliferation in late stages of retinal histogenesis, (2) EGF is mitogenic for Müller glia in vivo during the first two postnatal weeks in the rodent retina, (3) the effectiveness of EGF as a Müller glial mitogen declines in parallel with the decline in EGFR expression as the retina matures, and (4) following damage to the retina from continuous light exposure, EGFR expression is up-regulated in Müller glia to levels close to those in the neonatal retina, resulting in a renewed mitotic response to EGF. Together with previous results from other studies, these data indicate that the downregulation of a growth factor receptor is one mechanism by which glial cells maintain mitotic quiescence in the mature nervous system.


Assuntos
Proliferação de Células , Receptores ErbB/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Retina/citologia , Retina/metabolismo , Animais , Animais Recém-Nascidos , Receptores ErbB/genética , Receptores ErbB/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Estimulação Luminosa/métodos , Ratos , Retina/crescimento & desenvolvimento
17.
Neoplasia ; 7(9): 824-37, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16229805

RESUMO

TWIST, a basic helix-loop-helix (bHLH) transcription factor that regulates mesodermal development, has been shown to promote tumor cell metastasis and to enhance survival in response to cytotoxic stress. Our analysis of rat C6 glioma cell-derived cDNA revealed TWIST expression, suggesting that the gene may play a role in the genesis and physiology of primary brain tumors. To further delineate a possible oncogenic role for TWIST in the central nervous system (CNS), we analyzed TWIST expression in human gliomas and normal brain by using reverse transcription polymerase chain reaction, Northern blot analysis, in situ hybridization, and immunohistochemistry. TWIST expression was detected in the large majority of human glioma-derived cell lines and human gliomas examined. Levels of TWIST mRNA were associated with the highest grade gliomas, and increased TWIST expression accompanied transition from low grade to high grade in vivo, suggesting a role for TWIST in promoting malignant progression. In accord, elevated TWIST mRNA abundance preceded the spontaneous malignant transformation of cultured mouse astrocytes hemizygous for p53. Overexpression of TWIST protein in a human glioma cell line significantly enhanced tumor cell invasion, a hallmark of high-grade gliomas. These findings support roles for TWIST both in early glial tumorigenesis and subsequent malignant progression. TWIST was also expressed in embryonic and fetal human brain, and in neurons, but not glia, of mature brain, indicating that, in gliomas, TWIST may promote the functions also critical for CNS development or normal neuronal physiology.


Assuntos
Neoplasias do Sistema Nervoso Central/metabolismo , Glioma/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Astrocitoma/metabolismo , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Linhagem Celular Tumoral , Neoplasias do Sistema Nervoso Central/classificação , Neoplasias do Sistema Nervoso Central/patologia , Feto/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/classificação , Glioma/patologia , Humanos , Camundongos , Invasividade Neoplásica , Neurônios/química , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Fenótipo , RNA Mensageiro/metabolismo , Proteína 1 Relacionada a Twist/análise , Proteína 1 Relacionada a Twist/genética
18.
Dev Dyn ; 229(3): 529-40, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14991709

RESUMO

The ciliary epithelium of the ciliary body is derived from the anterior rim of the developing optic cup. Several recent studies have found that developmental abnormalities in this tissue can underlie congenital glaucoma. However, there is little known about the development of the ciliary epithelium. To better understand the developmental events responsible for the specification of this domain of the optic cup, we used a subtractive library, differential screening approach along with the construction of cDNA arrays to identify genes expressed in the ciliary epithelium of the chicken. We identified many genes specifically expressed in the ciliary epithelium, including a number that had been described previously as enriched in the ciliary epithelium of other species. By analyzing the expression of these genes during eye development, we were able to correlate the onset of ciliary epithelial gene expression with a reduction in mitotic activity in this region. We propose that the mechanisms that regulate the expression of ciliary epithelial genes are linked to the reduction in proliferation that results in the epithelial monolayer in this region.


Assuntos
Corpo Ciliar/embriologia , Animais , Divisão Celular , Galinhas , Colágeno Tipo IX/metabolismo , Cistatinas/biossíntese , DNA Complementar/metabolismo , Olho/embriologia , Biblioteca Gênica , Imuno-Histoquímica , Hibridização In Situ , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
19.
Dev Dyn ; 229(3): 555-63, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14991711

RESUMO

Müller glia have been shown to be a potential source of neural regeneration in the avian retina. In response to acute damage Müller glia de-differentiate, proliferate, express transcription factors found in embryonic retinal progenitors, and some of the progeny differentiate into neurons and glia (Fischer and Reh [2001a] Nat. Neurosci. 4:247-252). However, most of the cells produced by proliferating Müller cells appear to remain undifferentiated. The purpose of this study was to test whether the neurogenic gene NeuroD can promote the differentiation of proliferating cells derived from the postnatal chick retina. We used recombinant avian retroviruses to transfect green fluorescent protein (GFP) or NeuroD. The majority of cells transfected with GFP remained undifferentiated, with a few cells differentiating into calretinin-immunoreactive neurons. Many cells transfected with the NeuroD-virus expressed calretinin, neurofilament, or visinin, while most cells remained undifferentiated. The number of calretinin-expressing cells that were generated was increased approximately 20-fold with forced expression of NeuroD. In addition, we found that cells transfected with NeuroD never expressed glutamine synthetase, a marker of mature Müller glia, suggesting that NeuroD suppresses glial differentiation. We conclude that NeuroD stimulates cells from the toxin-damaged chicken retina to acquire some neuronal phenotypes. We propose that most of these cells were derived from Müller glia.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/fisiologia , Retina/embriologia , Proteína G de Ligação ao Cálcio S100/biossíntese , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Calbindina 2 , Diferenciação Celular , Divisão Celular , Embrião de Galinha , Galinhas , Glutamato-Amônia Ligase/biossíntese , Proteínas de Fluorescência Verde , Imuno-Histoquímica , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fenótipo , Retroviridae/genética , Fatores de Tempo , Toxinas Biológicas/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA