Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Nat Methods ; 15(5): 330-338, 2018 05.
Article in English | MEDLINE | ID: mdl-29638227

ABSTRACT

A key component of efforts to address the reproducibility crisis in biomedical research is the development of rigorously validated and renewable protein-affinity reagents. As part of the US National Institutes of Health (NIH) Protein Capture Reagents Program (PCRP), we have generated a collection of 1,406 highly validated immunoprecipitation- and/or immunoblotting-grade mouse monoclonal antibodies (mAbs) to 737 human transcription factors, using an integrated production and validation pipeline. We used HuProt human protein microarrays as a primary validation tool to identify mAbs with high specificity for their cognate targets. We further validated PCRP mAbs by means of multiple experimental applications, including immunoprecipitation, immunoblotting, chromatin immunoprecipitation followed by sequencing (ChIP-seq), and immunohistochemistry. We also conducted a meta-analysis that identified critical variables that contribute to the generation of high-quality mAbs. All validation data, protocols, and links to PCRP mAb suppliers are available at http://proteincapture.org.


Subject(s)
Antibodies, Monoclonal/immunology , Protein Array Analysis/methods , Transcription Factors/metabolism , Animals , Cloning, Molecular , Databases, Factual , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Reproducibility of Results
2.
Mol Ther ; 26(6): 1581-1593, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29673930

ABSTRACT

Most genetically distinct inherited retinal degenerations are primary photoreceptor degenerations. We selected a severe early onset form of Leber congenital amaurosis (LCA), caused by mutations in the gene LCA5, in order to test the efficacy of gene augmentation therapy for a ciliopathy. The LCA5-encoded protein, Lebercilin, is essential for the trafficking of proteins and vesicles to the photoreceptor outer segment. Using the AAV serotype AAV7m8 to deliver a human LCA5 cDNA into an Lca5 null mouse model of LCA5, we show partial rescue of retinal structure and visual function. Specifically, we observed restoration of rod-and-cone-driven electroretinograms in about 25% of injected eyes, restoration of pupillary light responses in the majority of treated eyes, an ∼20-fold decrease in target luminance necessary for visually guided behavior, and improved retinal architecture following gene transfer. Using LCA5 patient-derived iPSC-RPEs, we show that delivery of the LCA5 cDNA restores lebercilin protein and rescues cilia quantity. The results presented in this study support a path forward aiming to develop safety and efficacy trials for gene augmentation therapy in human subjects with LCA5 mutations. They also provide the framework for measuring the effects of intervention in ciliopathies and other severe, early-onset blinding conditions.


Subject(s)
Blindness/metabolism , Blindness/therapy , Dependovirus/genetics , Genetic Therapy/methods , Animals , Electroretinography , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/therapy , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
3.
Development ; 145(9)2018 04 30.
Article in English | MEDLINE | ID: mdl-29650591

ABSTRACT

Precise control of the relative ratio of retinal neurons and glia generated during development is essential for visual function. We show that Lhx2, which encodes a LIM-homeodomain transcription factor essential for specification and differentiation of retinal Müller glia, also plays a crucial role in the development of retinal neurons. Overexpression of Lhx2 with its transcriptional co-activator Ldb1 triggers cell cycle exit and inhibits both Notch signaling and retinal gliogenesis. Lhx2/Ldb1 overexpression also induces the formation of wide-field amacrine cells (wfACs). In contrast, Rnf12, which encodes a negative regulator of LDB1, is necessary for the initiation of retinal gliogenesis. We also show that Lhx2-dependent neurogenesis and wfAC formation requires Ascl1 and Neurog2, and that Lhx2 is necessary for their expression, although overexpression of Lhx2/Ldb1 does not elevate expression of these proneural bHLH factors. Finally, we demonstrate that the relative level of the LHX2-LDB1 complex in the retina decreases in tandem with the onset of gliogenesis. These findings show that control of Lhx2 function by Ldb1 and Rnf12 underpins the coordinated differentiation of neurons and Müller glia in postnatal retina.


Subject(s)
DNA-Binding Proteins/metabolism , Ependymoglial Cells/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Neurogenesis/physiology , Retinal Neurons/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/genetics , Ependymoglial Cells/cytology , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retinal Neurons/cytology , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics
4.
Methods Mol Biol ; 1715: 101-111, 2018.
Article in English | MEDLINE | ID: mdl-29188509

ABSTRACT

In vivo electroporation enables the transformation of retinal tissue with engineered DNA plasmids, facilitating the selective expression of desired gene products. This method achieves plasmid transfer via the application of an external electrical field, which both generates a transient increase in the permeability of cell plasma membranes, and promotes the incorporation of DNA plasmids by electrophoretic transfer through the permeabilized membranes. Here we describe a method for the preparation, injection, and electroporation of DNA plasmids into neonatal mouse retinal tissue. This method can be utilized to perform gain of function or loss of function studies in the mouse. Experimental design is limited only by construct availability.


Subject(s)
Electroporation/methods , Gene Transfer Techniques , Plasmids , Retina/physiology , Animals , Animals, Newborn , DNA/administration & dosage , DNA/genetics , Mice , Retina/growth & development
5.
Development ; 144(9): 1698-1711, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28356311

ABSTRACT

Regulated retinal ganglion cell (RGC) differentiation and axonal guidance is required for a functional visual system. Homeodomain and basic helix-loop-helix transcription factors are required for retinogenesis, as well as patterning, differentiation and maintenance of specific retinal cell types. We hypothesized that Dlx1, Dlx2 and Brn3b homeobox genes function in parallel intrinsic pathways to determine RGC fate and therefore generated Dlx1/Dlx2/Brn3b triple-knockout mice. A more severe retinal phenotype was found in the Dlx1/Dlx2/Brn3b-null retinas than was predicted by combining features of the Brn3b single- and Dlx1/Dlx2 double-knockout retinas, including near total RGC loss with a marked increase in amacrine cells in the ganglion cell layer. Furthermore, we discovered that DLX1 and DLX2 function as direct transcriptional activators of Brn3b expression. Knockdown of Dlx2 expression in primary embryonic retinal cultures and Dlx2 gain of function in utero strongly support that DLX2 is both necessary and sufficient for Brn3b expression in vivo We suggest that ATOH7 specifies RGC-committed progenitors and that Dlx1 and Dlx2 function both downstream of ATOH7 and in parallel, but cooperative, pathways that involve regulation of Brn3b expression to determine RGC fate.


Subject(s)
Cell Differentiation , Homeodomain Proteins/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Transcription Factor Brn-3B/metabolism , Transcription Factors/metabolism , Vertebrates/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Apoptosis/genetics , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Division/genetics , Cell Lineage/genetics , Cell Proliferation , Cells, Cultured , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Electroporation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice, Knockout , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor Brn-3B/deficiency , Transcription Factors/deficiency
6.
Development ; 143(22): 4182-4192, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27697904

ABSTRACT

The Lim domain-binding proteins are key co-factor proteins that assemble with LIM domains of the LMO/LIM-HD family to form functional complexes that regulate cell proliferation and differentiation. Using conditional mutagenesis and comparative phenotypic analysis, we analyze the function of Ldb1 and Ldb2 in mouse retinal development, and demonstrate overlapping and specific functions of both proteins. Ldb1 interacts with Lhx2 in the embryonic retina and both Ldb1 and Ldb2 play a key role in maintaining the pool of retinal progenitor cells. This is accomplished by controlling the expression of the Vsx2 and Rax, and components of the Notch and Hedgehog signaling pathways. Furthermore, the Ldb1/Ldb2-mediated complex is essential for generation of early-born photoreceptors through the regulation of Rax and Crx. Finally, we demonstrate functional redundancy between Ldb1 and Ldb2. Ldb1 can fully compensate the loss of Ldb2 during all phases of retinal development, whereas Ldb2 alone is sufficient to sustain activity of Lhx2 in both early- and late-stage RPCs and in Müller glia. By contrast, loss of Ldb1 disrupts activity of the LIM domain factors in neuronal precursors. An intricate regulatory network exists that is mediated by Ldb1 and Ldb2, and promotes RPC proliferation and multipotency; it also controls specification of mammalian retina cells.


Subject(s)
DNA-Binding Proteins/physiology , LIM Domain Proteins/physiology , Organogenesis/genetics , Retina/embryology , Transcription Factors/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Gene Regulatory Networks/physiology , Mammals/embryology , Mammals/genetics , Mice , Mice, Transgenic , Retina/cytology , Retina/metabolism , Stem Cells/physiology
7.
Development ; 143(21): 3994-4002, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27633990

ABSTRACT

Fibroblast growth factor (FGF) signaling is an essential regulator of lens epithelial cell proliferation and survival, as well as lens fiber cell differentiation. However, the identities of these FGF factors, their source tissue and the genes that regulate their synthesis are unknown. We have found that Chx10-Cre;Lhx2lox/lox mice, which selectively lack Lhx2 expression in neuroretina from E10.5, showed an early arrest in lens fiber development along with severe microphthalmia. These mutant animals showed reduced expression of multiple neuroretina-expressed FGFs and canonical FGF-regulated genes in neuroretina. When FGF expression was genetically restored in Lhx2-deficient neuroretina of Chx10-Cre;Lhx2lox/lox mice, we observed a partial but nonetheless substantial rescue of the defects in lens cell proliferation, survival and fiber differentiation. These data demonstrate that neuroretinal expression of Lhx2 and neuroretina-derived FGF factors are crucial for lens fiber development in vivo.


Subject(s)
Fibroblast Growth Factors/genetics , LIM-Homeodomain Proteins/physiology , Lens, Crystalline/embryology , Organogenesis/genetics , Retinal Neurons/physiology , Transcription Factors/physiology , Animals , Cell Differentiation/genetics , Embryo, Mammalian , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/genetics , Lens, Crystalline/metabolism , Mice , Mice, Transgenic , Microphthalmos/embryology , Microphthalmos/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Retinal Neurons/metabolism , Signal Transduction/genetics , Transcription Factors/genetics
8.
Sci Rep ; 6: 32757, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27605455

ABSTRACT

Müller glia (MG) are the principal glial cell type in the vertebrate retina. Recent work has identified the LIM homeodomain factor encoding gene Lhx2 as necessary for both Notch signaling and MG differentiation in late-stage retinal progenitor cells (RPCs). However, the extent to which Lhx2 interacts with other intrinsic regulators of MG differentiation is unclear. We investigated this question by investigating the effects of overexpression of multiple transcriptional regulators that are either known or hypothesized to control MG formation, in both wildtype and Lhx2-deficient RPCs. We observe that constitutively elevated Notch signaling, induced by N1ICD electroporation, inhibited gliogenesis in wildtype animals, but rescued MG development in Lhx2-deficient retinas. Electroporation of Nfia promoted the formation of cells with MG-like radial morphology, but did not drive expression of MG molecular markers. Plagl1 and Sox9 did not induce gliogenesis in wildtype animals, but nonetheless activated expression of the Müller marker P27(Kip1) in Lhx2-deficient cells. Finally, Sox2, Sox8, and Sox9 promoted amacrine cell formation in Lhx2-deficient cells, but not in wildtype retinas. These findings demonstrate that overexpression of individual gliogenic factors typically regulates only a subset of characteristic MG markers, and that these effects are differentially modulated by Lhx2.


Subject(s)
LIM-Homeodomain Proteins/metabolism , Neuroglia/physiology , Retina/cytology , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Electroporation , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Gene Expression Regulation , Genes, Tumor Suppressor , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Male , Mice, Inbred Strains , Mice, Knockout , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Neuroepithelial Cells/physiology , Neuroglia/cytology , Retina/metabolism , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Transcription Factors/genetics
9.
J Neurosci ; 36(8): 2391-405, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26911688

ABSTRACT

Müller glia (MG) are the only glial cell type produced by the neuroepithelial progenitor cells that generate the vertebrate retina. MG are required to maintain retinal homeostasis and support the survival of retinal neurons. Furthermore, in certain vertebrate classes, MG function as adult stem cells, mediating retinal regeneration in response to injury. However, the mechanisms that regulate MG development are poorly understood because there is considerable overlap in gene expression between retinal progenitor cells and differentiated MG. We show that the LIM homeodomain transcription factor Lhx2 is required for the development of MG in the mouse retina. Temporally controlled knock-out studies reveal a requirement for Lhx2 during all stages of MG development, ranging from the proliferation of gliocompetent retinal progenitors, activation of Müller-specific gene expression, and terminal differentiation of MG morphological features. We show that Lhx2 regulates gliogenesis in part by regulating directly the expression of Notch pathway genes including Notch1, Dll1, and Dll3 and gliogenic transcription factors such as Hes1, Hes5, Sox8, and Rax. Conditional knock-out of Lhx2 resulted in a rapid downregulation of Notch pathway genes and loss of Notch signaling. We further demonstrate that Müller gliogenesis induced by misexpression of the potently gliogenic Notch pathway transcriptional effector Hes5 requires Lhx2 expression. These results indicate that Lhx2 not only directly regulates expression of Notch signaling pathway components, but also acts together with the gliogenic Notch pathway to drive MG specification and differentiation.


Subject(s)
LIM-Homeodomain Proteins/biosynthesis , Neuroglia/metabolism , Receptor, Notch1/biosynthesis , Retinal Neurons/metabolism , Signal Transduction/physiology , Transcription Factors/biosynthesis , Animals , Animals, Newborn , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Retina
10.
J Neurosci ; 34(50): 16809-20, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505333

ABSTRACT

Hypothalamic tanycytes, a radial glial-like ependymal cell population that expresses numerous genes selectively enriched in embryonic hypothalamic progenitors and adult neural stem cells, have recently been observed to serve as a source of adult-born neurons in the mammalian brain. The genetic mechanisms that regulate the specification and maintenance of tanycyte identity are unknown, but are critical for understanding how these cells can act as adult neural progenitor cells. We observe that LIM (Lin-11, Isl-1, Mec-3)-homeodomain gene Lhx2 is selectively expressed in hypothalamic progenitor cells and tanycytes. To test the function of Lhx2 in tanycyte development, we used an intersectional genetic strategy to conditionally delete Lhx2 in posteroventral hypothalamic neuroepithelium, both embryonically and postnatally. We observed that tanycyte development was severely disrupted when Lhx2 function was ablated during embryonic development. Lhx2-deficient tanycytes lost expression of tanycyte-specific genes, such as Rax, while also displaying ectopic expression of genes specific to cuboid ependymal cells, such as Rarres2. Ultrastructural analysis revealed that mutant tanycytes exhibited a hybrid identity, retaining radial morphology while becoming multiciliated. In contrast, postnatal loss of function of Lhx2 resulted only in loss of expression of tanycyte-specific genes. Using chromatin immunoprecipitation, we further showed that Lhx2 directly regulated expression of Rax, an essential homeodomain factor for tanycyte development. This study identifies Lhx2 as a key intrinsic regulator of tanycyte differentiation, sustaining Rax-dependent activation of tanycyte-specific genes while also inhibiting expression of ependymal cell-specific genes. These findings provide key insights into the transcriptional regulatory network specifying this still poorly characterized cell type.


Subject(s)
Cell Differentiation/physiology , Ependymoglial Cells/physiology , Hypothalamus/cytology , Hypothalamus/physiology , LIM-Homeodomain Proteins/physiology , Neurogenesis/physiology , Transcription Factors/physiology , Animals , Female , Male , Mice , Mice, Transgenic
11.
J Neurosci ; 33(16): 6877-84, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23595746

ABSTRACT

Eye formation is regulated by a complex network of eye field transcription factors (EFTFs), including LIM-homeodomain gene LHX2. We disrupted LHX2 function at different stages during this process using a conditional knock-out strategy in mice. We find that LHX2 function is required in an ongoing fashion to maintain optic identity across multiple stages, from the formation of the optic vesicle to the differentiation of the neuroretina. At each stage, loss of Lhx2 led to upregulation of a set of molecular markers that are normally expressed in the thalamic eminence and in the anterodorsal hypothalamus in a portion of the optic vesicle or retina. Furthermore, the longer LHX2 function was maintained, the further optic morphogenesis progressed. Early loss of function caused profound mispatterning of the entire telencephalic-optic-hypothalamic field, such that the optic vesicle became mispositioned and appeared to arise from the diencephalic-telencephalic boundary. At subsequent stages, loss of Lhx2 did not affect optic vesicle position but caused arrest of optic cup formation. If Lhx2 was selectively disrupted in the neuroretina from E11.5, the neuroretina showed gross dysmorphology along with aberrant expression of markers specific to the thalamic eminence and anterodorsal hypothalamus. Our findings indicate a continual requirement for LHX2 throughout the early stages of optic development, not only to maintain optic identity by suppressing alternative fates but also to mediate multiple steps of optic morphogenesis. These findings provide new insight into the anophthalmic phenotype of the Lhx2 mutant and reveal novel roles for this transcription factor in eye development.


Subject(s)
Gene Expression Regulation, Developmental/genetics , LIM-Homeodomain Proteins/metabolism , Morphogenesis/genetics , Organogenesis/genetics , Transcription Factors/metabolism , Visual Pathways/physiology , Age Factors , Animals , Body Patterning/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Proteins/genetics , RNA, Untranslated , Repressor Proteins/metabolism , Retina/abnormalities , Retina/pathology , Tamoxifen/pharmacology , Transcription Factors/genetics , Visual Pathways/embryology
12.
Proc Natl Acad Sci U S A ; 109(12): 4657-62, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22393024

ABSTRACT

Müller glia are the primary glial subtype in the retina and perform a wide range of physiological tasks in support of retinal function, but little is known about the transcriptional network that maintains these cells in their differentiated state. We report that selective deletion of the LIM homeodomain transcription factor Lhx2 from mature Müller glia leads to the induction of reactive retinal gliosis in the absence of injury. Furthermore, Lhx2 expression is also down-regulated in Prph2(Rd2/Rd2) animals immediately before the onset of reactive gliosis. Analysis of conditional Lhx2 knockouts showed that gliosis was hypertrophic but not proliferative. Aging of experimental animals demonstrated that constitutive reactive gliosis induced by deletion of Lhx2 reduced rates of ongoing apoptosis and compromised both rod and cone photoreceptor function. Additionally, these animals showed a dramatically reduced ability to induce expression of secreted neuroprotective factors and displayed enhanced rates of apoptosis in light-damage assays. We provide in vivo evidence that Lhx2 actively maintains mature Müller glia in a nonreactive state, with loss of function initiating a specific program of nonproliferative hypertrophic gliosis.


Subject(s)
Gliosis/pathology , LIM-Homeodomain Proteins/physiology , Retina/pathology , Transcription Factors/physiology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Gene Expression Regulation , Gliosis/genetics , LIM-Homeodomain Proteins/genetics , Light , Mice , Mice, Knockout , Models, Biological , Neurodegenerative Diseases/metabolism , Neuroglia/physiology , Protein Structure, Tertiary , Retina/physiology , Tamoxifen/pharmacology , Transcription Factors/genetics
13.
J Vis Exp ; (52)2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21730944

ABSTRACT

The functional characterization of genes expressed during mammalian retinal development remains a significant challenge. Gene targeting to generate constitutive or conditional loss of function knockouts remains cost and labor intensive, as well as time consuming. Adding to these challenges, retina expressed genes may have essential roles outside the retina leading to unintended confounds when using a knockout approach. Furthermore, the ability to ectopically express a gene in a gain of function experiment can be extremely valuable when attempting to identify a role in cell fate specification and/or terminal differentiation. We present a method for the rapid and efficient incorporation of DNA plasmids into the neonatal mouse retina by electroporation. The application of short electrical impulses above a certain field strength results in a transient increase in plasma membrane permeability, facilitating the transfer of material across the membrane (1,2,3,4). Groundbreaking work demonstrated that electroporation could be utilized as a method of gene transfer into mammalian cells by inducing the formation of hydrophilic plasma membrane pores allowing the passage of highly charged DNA through the lipid bilayer (5). Continuous technical development has resulted in the viability of electroporation as a method for in vivo gene transfer in multiple mouse tissues including the retina, the method for which is described herein (6, 7, 8, 9, 10). DNA solution is injected into the subretinal space so that DNA is placed between the retinal pigmented epithelium and retina of the neonatal (P0) mouse and electrical pulses are applied using a tweezer electrode. The lateral placement of the eyes in the mouse allows for the easy orientation of the tweezer electrode to the necessary negative pole-DNA-retina-positive pole alignment. Extensive incorporation and expression of transferred genes can be identified by postnatal day 2 (P2). Due to the lack of significant lateral migration of cells in the retina, electroporated and non-electroporated regions are generated. Non-electroporated regions may serve as internal histological controls where appropriate. Retinal electroporation can be used to express a gene under a ubiquitous promoter, such as CAG, or to disrupt gene function using shRNA constructs or Cre-recombinase. More targeted expression can be achieved by designing constructs with cell specific gene promoters. Visualization of electroporated cells is achieved using bicistronic constructs expressing GFP or by co-electroporating a GFP expression construct. Furthermore, multiple constructs may be electroporated for the study of combinatorial gene effects or simultaneous gain and loss of function of different genes. Retinal electroporation may also be utilized for the analysis of genomic cis-regulatory elements by generating appropriate expression constructs and deletion mutants. Such experiments can be used to identify cis-regulatory regions sufficient or required for cell specific gene expression (11). Potential experiments are limited only by construct availability.


Subject(s)
DNA/administration & dosage , Electroporation/methods , Plasmids/administration & dosage , Retina/physiology , Animals , DNA/genetics , Mice , Plasmids/genetics
14.
Development ; 138(11): 2325-36, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558380

ABSTRACT

The mammalian retina is a tractable model system for analyzing transcriptional networks that guide neural development. Spalt family zinc-finger transcription factors play a crucial role in photoreceptor specification in Drosophila, but their role in mammalian retinal development has not been investigated. In this study, we show that that the spalt homolog Sall3 is prominently expressed in developing cone photoreceptors and horizontal interneurons of the mouse retina and in a subset of cone bipolar cells. We find that Sall3 is both necessary and sufficient to activate the expression of multiple cone-specific genes, and that Sall3 protein is selectively bound to the promoter regions of these genes. Notably, Sall3 shows more prominent expression in short wavelength-sensitive cones than in medium wavelength-sensitive cones, and that Sall3 selectively activates expression of the short but not the medium wavelength-sensitive cone opsin gene. We further observe that Sall3 regulates the differentiation of horizontal interneurons, which form direct synaptic contacts with cone photoreceptors. Loss of function of Sall3 eliminates expression of the horizontal cell-specific transcription factor Lhx1, resulting in a radial displacement of horizontal cells that partially phenocopies the loss of function of Lhx1. These findings not only demonstrate that Spalt family transcription factors play a conserved role in regulating photoreceptor development in insects and mammals, but also identify Sall3 as a factor that regulates terminal differentiation of both cone photoreceptors and their postsynaptic partners.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Horizontal Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Chromatin Immunoprecipitation , Electroporation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , In Situ Nick-End Labeling , LIM-Homeodomain Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Opsins/genetics , Promoter Regions, Genetic , Retina/cytology , Retina/growth & development , Retina/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Horizontal Cells/cytology , Transcription Factors/biosynthesis
15.
Proc Natl Acad Sci U S A ; 107(25): 11579-84, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20534447

ABSTRACT

Mutation of rod photoreceptor-enriched transcription factors is a major cause of inherited blindness. We identified the orphan nuclear hormone receptor estrogen-related receptor beta (ERRbeta) as selectively expressed in rod photoreceptors. Overexpression of ERRbeta induces expression of rod-specific genes in retinas of wild-type as well as Nrl(-/-) mice, which lack rod photoreceptors. Mutation of ERRbeta results in dysfunction and degeneration of rods, whereas inverse agonists of ERRbeta trigger rapid rod degeneration, which is rescued by constitutively active mutants of ERRbeta. ERRbeta coordinates expression of multiple genes that are rate-limiting regulators of ATP generation and consumption in photoreceptors. Furthermore, enhancing ERRbeta activity rescues photoreceptor defects that result from loss of the photoreceptor-specific transcription factor Crx. Our findings demonstrate that ERRbeta is a critical regulator of rod photoreceptor function and survival, and suggest that ERRbeta agonists may be useful in the treatment of certain retinal dystrophies.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Receptors, Estrogen/metabolism , Retina/embryology , Retinal Rod Photoreceptor Cells/metabolism , Trans-Activators/physiology , Animals , Cell Survival , Electroretinography/methods , Homeodomain Proteins/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhodopsin/metabolism , Trans-Activators/metabolism
16.
Nucleic Acids Res ; 36(3): 872-84, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18086710

ABSTRACT

Dlx homeobox genes are first expressed in embryonic retina at E11.5. The Dlx1/Dlx2 null retina has a reduced ganglion cell layer (GCL), with loss of late-born differentiated retinal ganglion cells (RGCs) due to increased apoptosis. TrkB signaling is proposed to regulate the dynamics of RGC apoptosis throughout development. DLX2 expression markedly precedes the onset of TrkB expression in the GCL; TrkB co-expression with Dlx2 and RGC markers is well-established by E13.5. In the Dlx1/Dlx2 null retina, TrkB expression is significantly reduced by E16.5. We demonstrated that DLX2 binds to a specific region of the TrkB promoter in retinal neuroepithelium during embryogenesis. In vitro confirmation and the functional consequences of DLX2 binding to this TrkB regulatory region support TrkB as a Dlx2 transcriptional target. Furthermore, ectopic Dlx2 expression in retinal explants activates TrkB expression and Dlx2 knockdown in primary retinal cultures results in reduced TrkB expression. RGC differentiation and survival require the coordinated expression of transcription factors. This study establishes a direct transcriptional relationship between a homeodomain protein involved in RGC differentiation and a neurotrophin receptor implicated in RGC survival. Signaling mediated by TrkB may contribute to survival of late-born RGCs whose terminal differentiation is regulated by Dlx gene function.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Membrane Glycoproteins/genetics , Protein-Tyrosine Kinases/genetics , Retina/embryology , Retinal Ganglion Cells/metabolism , Transcription Factors/metabolism , Amacrine Cells/metabolism , Animals , Base Sequence , Binding Sites , Cells, Cultured , Enhancer Elements, Genetic , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Protein-Tyrosine Kinases/metabolism , RNA Interference , Retina/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Activation
17.
Arch Immunol Ther Exp (Warsz) ; 53(4): 308-20, 2005.
Article in English | MEDLINE | ID: mdl-16088315

ABSTRACT

In this review we focus on peptide- and peptidomimetic-based approaches that target autoimmune diseases and some pathologies of the central nervous system. Special attention is given to asthma, allergic rhinitis, osteoarthritis, and Alzheimer's disease, but other related pathologies are also reviewed, although to a lesser degree. Among others, drugs like Diacerhein and its active form Rhein, Pralnacasan, Anakinra (Kineret), Omalizumab, an antibody "BION-1", directed against the common beta-chain of cytokine receptors, are described below as well as attempts to target beta-amyloid peptide aggregation. Parts of the review are also dedicated to targeting of pathologic conditions in the brain and in other tissues with peptides as well as methods to deliver larger molecules through the "blood--brain barrier" by exploring receptor-mediated transport, or elsewhere in the body by using peptides as carriers through cellular membranes. In addition to highlighting current developments in the field, we also propose, for future drug targets, the components of the inflammasome protein complex, which is believed to initiate the activation of caspase- 1 dependent signaling events, as well as other pathways that signal inflammation. Thus we discuss the possibility of targeting inflammasome components for negative or positive modulation of an inflammatory response.


Subject(s)
Arthritis/therapy , Asthma/therapy , Autoimmune Diseases/therapy , Central Nervous System Diseases/therapy , Peptides/therapeutic use , Amyloid beta-Peptides/chemistry , Animals , Anti-Asthmatic Agents/pharmacology , Cell Membrane/metabolism , Humans , Inflammation , Models, Biological , Peptides/chemistry
18.
Development ; 132(2): 311-22, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15604100

ABSTRACT

Dlx homeobox genes, the vertebrate homologs of Distal-less, play important roles in the development of the vertebrate forebrain, craniofacial structures and limbs. Members of the Dlx gene family are also expressed in retinal ganglion cells (RGC), amacrine and horizontal cells of the developing and postnatal retina. Expression begins at embryonic day 12.5 and is maintained until late embryogenesis for Dlx1, while Dlx2 expression extends to adulthood. We have assessed the retinal phenotype of the Dlx1/Dlx2 double knockout mouse, which dies at birth. The Dlx1/2 null retina displays a reduced ganglion cell layer (GCL), with loss of differentiated RGCs due to increased apoptosis, and corresponding thinning of the optic nerve. Ectopic expression of Crx, the cone and rod photoreceptor homeobox gene, in the GCL and neuroblastic layers of the mutants may signify altered cell fate of uncommitted RGC progenitors. However, amacrine and horizontal cell differentiation is relatively unaffected in the Dlx1/2 null retina. Herein, we propose a model whereby early-born RGCs are Dlx1 and Dlx2 independent, but Dlx function is necessary for terminal differentiation of late-born RGC progenitors.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Retina/embryology , Retinal Ganglion Cells/metabolism , Animals , Apoptosis , Brain/metabolism , Bromodeoxyuridine/pharmacology , Cell Differentiation , Homeodomain Proteins/genetics , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Biological , Mutation , Phenotype , Retina/metabolism , Time Factors , Transcription Factors
19.
Nucleic Acids Res ; 32(3): 884-92, 2004.
Article in English | MEDLINE | ID: mdl-14769946

ABSTRACT

Understanding homeobox gene specificity and function has been hampered by the lack of proven direct transcriptional targets during development. Dlx genes are expressed in the developing forebrain, retina, craniofacial structures and limbs. Dlx1/Dlx2 double knockout mice die at birth with multiple defects including abnormal forebrain development and decreased Dlx5 and Dlx6 expression. We have successfully applied chromatin immunoprecipitation (ChIP) to identify a direct transcriptional target of DLX homeoproteins from embryonic tissues in vivo. We optimized cross-linking conditions to enrich for protein-DNA complexes, then using specific high affinity DLX antibodies captured immunoenriched DLX genomic DNA transcriptional targets. DLX homeobox proteins bind differentially to the Dlx5/Dlx6 intergenic enhancer in newborn retina (DLX2) and embryonic striatum (DLX1, DLX2) in situ. Reporter gene assays demonstrated the functional significance of the binding of DLX proteins to this regulatory element, confirmed in vitro by electrophoretic mobility shift assays, using tissue extracts or recombinant DLX proteins. ChIP provides the best approach to identify direct Dlx homeoprotein targets from developing tissues in situ. The use of this technology will advance our understanding of Dlx gene function in the vertebrate in vivo and can be applied to examine targets of other homeobox genes and other classes of transcription factors.


Subject(s)
Homeodomain Proteins/metabolism , Prosencephalon/embryology , Retina/embryology , Transcription Factors/metabolism , Animals , Binding Sites , Chromatin/metabolism , Cisplatin/chemistry , Cross-Linking Reagents , DNA/metabolism , DNA, Intergenic/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Mice , Neostriatum/metabolism , Nuclear Proteins/metabolism , Precipitin Tests , Prosencephalon/metabolism , RNA, Messenger/metabolism , Retina/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transcriptional Activation
20.
J Comp Neurol ; 461(2): 187-204, 2003 Jun 23.
Article in English | MEDLINE | ID: mdl-12724837

ABSTRACT

Distal-less homeobox genes are expressed in the developing forebrain. We assessed Dlx gene expression in the developing and adult mouse retina. Dlx1 and Dlx2 are detected in retinal neuroprogenitors by embryonic day (E) 12.5 (Eisenstat et al. [1999] J. Comp. Neurol. 217-237). At E13.5, the expression of four homeodomain proteins, DLX2, BRN3b, PAX6, and CHX10, define distinct yet overlapping domains in the retinal neuroepithelium. By postnatal day (P) 0, DLX2 is expressed in the neuroblastic layer and the ganglion cell layer (GCL) consisting of ganglion and displaced amacrine cells. DLX1 expression resembles DLX2 to P0 but decreases postnatally. In the adult, DLX2 is localized to ganglion, amacrine, and horizontal cells as determined by coexpression with retinal cell-specific markers. There is coincident expression of DLX2 with gamma-aminobutyric acid (GABA), glutamic acid decarboxylase (GAD)65, and GAD67 in the inner nuclear layer (INL) and GCL. In the adult, DLX2 is coexpressed with BRN3b in ganglion cells; PAX6 in amacrine, horizontal, and ganglion cells; and Chx10 in some bipolar cells. We predict that a combinatorial code of these homeobox genes and others specify retinal cell fate. Our results support a possible role for Dlx1 and Dlx2 in inner retinal development and in the terminal differentiation and/or maintenance of INL interneurons and ganglion cells in the adult. The correlation of DLX2 with GABA expression in the mouse retina closely mirrors the relationship of DLX2 to GABAergic neuronal differentiation in the embryonic forebrain, including neocortex, olfactory bulb and hippocampus, signifying a conservation of function of Dlx genes in the developing central nervous system.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Retina/embryology , Retinal Ganglion Cells/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Cell Lineage/genetics , Cytoskeletal Proteins , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dopamine/metabolism , Eye Proteins , Fetus , Glutamate Decarboxylase/metabolism , Homeodomain Proteins/genetics , Immunohistochemistry , Interneurons/cytology , Interneurons/metabolism , Isoenzymes/metabolism , Mice , Mice, Inbred ICR , PAX6 Transcription Factor , Paired Box Transcription Factors , RNA, Messenger/metabolism , RNA-Binding Proteins , Repressor Proteins , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/cytology , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factor Brn-3 , Transcription Factor Brn-3B , Transcription Factors/genetics , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...