Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Genes Dev ; 33(15-16): 983-1007, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31123060

ABSTRACT

All transcription factors are equal, but some are more equal than others. In the 25 yr since the gene encoding the microphthalmia-associated transcription factor (MITF) was first isolated, MITF has emerged as a key coordinator of many aspects of melanocyte and melanoma biology. Like all transcription factors, MITF binds to specific DNA sequences and up-regulates or down-regulates its target genes. What marks MITF as being remarkable among its peers is the sheer range of biological processes that it appears to coordinate. These include cell survival, differentiation, proliferation, invasion, senescence, metabolism, and DNA damage repair. In this article we present our current understanding of MITF's role and regulation in development and disease, as well as those of the MITF-related factors TFEB and TFE3, and highlight key areas where our knowledge of MITF regulation and function is limited.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Melanocytes/physiology , Melanoma/physiopathology , Microphthalmia-Associated Transcription Factor/metabolism , Animals , Genome , Humans , Microphthalmia-Associated Transcription Factor/genetics , Protein Binding , Protein Isoforms
2.
PLoS Genet ; 13(12): e1007093, 2017 12.
Article in English | MEDLINE | ID: mdl-29240767

ABSTRACT

Congenital nephron number varies widely in the human population and individuals with low nephron number are at risk of developing hypertension and chronic kidney disease. The development of the kidney occurs via an orchestrated morphogenetic process where metanephric mesenchyme and ureteric bud reciprocally interact to induce nephron formation. The genetic networks that modulate the extent of this process and set the final nephron number are mostly unknown. Here, we identified a specific isoform of MITF (MITF-A), a bHLH-Zip transcription factor, as a novel regulator of the final nephron number. We showed that overexpression of MITF-A leads to a substantial increase of nephron number and bigger kidneys, whereas Mitfa deficiency results in reduced nephron number. Furthermore, we demonstrated that MITF-A triggers ureteric bud branching, a phenotype that is associated with increased ureteric bud cell proliferation. Molecular studies associated with an in silico analyses revealed that amongst the putative MITF-A targets, Ret was significantly modulated by MITF-A. Consistent with the key role of this network in kidney morphogenesis, Ret heterozygosis prevented the increase of nephron number in mice overexpressing MITF-A. Collectively, these results uncover a novel transcriptional network that controls branching morphogenesis during kidney development and identifies one of the first modifier genes of nephron endowment.


Subject(s)
Kidney/physiology , Microphthalmia-Associated Transcription Factor/metabolism , Nephrons/physiology , Animals , Female , Humans , Kidney/embryology , Kidney/metabolism , Male , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Morphogenesis , Nephrons/anatomy & histology , Nephrons/growth & development , Nephrons/metabolism , Organogenesis , Protein Isoforms , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Ureter/metabolism , Ureter/physiology
3.
Nature ; 469(7331): 548-53, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21248750

ABSTRACT

Cutaneous malignant melanoma is a highly aggressive and frequently chemoresistant cancer, the incidence of which continues to rise. Epidemiological studies show that the major aetiological melanoma risk factor is ultraviolet (UV) solar radiation, with the highest risk associated with intermittent burning doses, especially during childhood. We have experimentally validated these epidemiological findings using the hepatocyte growth factor/scatter factor transgenic mouse model, which develops lesions in stages highly reminiscent of human melanoma with respect to biological, genetic and aetiological criteria, but only when irradiated as neonatal pups with UVB, not UVA. However, the mechanisms underlying UVB-initiated, neonatal-specific melanomagenesis remain largely unknown. Here we introduce a mouse model permitting fluorescence-aided melanocyte imaging and isolation following in vivo UV irradiation. We use expression profiling to show that activated neonatal skin melanocytes isolated following a melanomagenic UVB dose bear a distinct, persistent interferon response signature, including genes associated with immunoevasion. UVB-induced melanocyte activation, characterized by aberrant growth and migration, was abolished by antibody-mediated systemic blockade of interferon-γ (IFN-γ), but not type-I interferons. IFN-γ was produced by macrophages recruited to neonatal skin by UVB-induced ligands to the chemokine receptor Ccr2. Admixed recruited skin macrophages enhanced transplanted melanoma growth by inhibiting apoptosis; notably, IFN-γ blockade abolished macrophage-enhanced melanoma growth and survival. IFN-γ-producing macrophages were also identified in 70% of human melanomas examined. Our data reveal an unanticipated role for IFN-γ in promoting melanocytic cell survival/immunoevasion, identifying a novel candidate therapeutic target for a subset of melanoma patients.


Subject(s)
Interferon-gamma/metabolism , Melanocytes/metabolism , Melanoma/physiopathology , Ultraviolet Rays , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/radiation effects , Humans , Macrophages/metabolism , Macrophages/radiation effects , Male , Melanocytes/radiation effects , Mice
4.
PLoS Genet ; 10(5): e1004360, 2014.
Article in English | MEDLINE | ID: mdl-24875170

ABSTRACT

During organogenesis, PAX6 is required for establishment of various progenitor subtypes within the central nervous system, eye and pancreas. PAX6 expression is maintained in a variety of cell types within each organ, although its role in each lineage and how it acquires cell-specific activity remain elusive. Herein, we aimed to determine the roles and the hierarchical organization of the PAX6-dependent gene regulatory network during the differentiation of the retinal pigmented epithelium (RPE). Somatic mutagenesis of Pax6 in the differentiating RPE revealed that PAX6 functions in a feed-forward regulatory loop with MITF during onset of melanogenesis. PAX6 both controls the expression of an RPE isoform of Mitf and synergizes with MITF to activate expression of genes involved in pigment biogenesis. This study exemplifies how one kernel gene pivotal in organ formation accomplishes a lineage-specific role during terminal differentiation of a single lineage.


Subject(s)
Cell Differentiation/genetics , Eye Proteins/biosynthesis , Homeodomain Proteins/biosynthesis , Microphthalmia-Associated Transcription Factor/genetics , Organogenesis/genetics , Paired Box Transcription Factors/biosynthesis , Repressor Proteins/biosynthesis , Animals , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Microphthalmia-Associated Transcription Factor/biosynthesis , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Pigmentation/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Retinal Pigment Epithelium/growth & development , Retinal Pigment Epithelium/metabolism
5.
PLoS Genet ; 8(7): e1002757, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22792072

ABSTRACT

The separation of the optic neuroepithelium into future retina and retinal pigment epithelium (RPE) is a critical event in early eye development in vertebrates. Here we show in mice that the transcription factor PAX6, well-known for its retina-promoting activity, also plays a crucial role in early pigment epithelium development. This role is seen, however, only in a background genetically sensitized by mutations in the pigment cell transcription factor MITF. In fact, a reduction in Pax6 gene dose exacerbates the RPE-to-retina transdifferentiation seen in embryos homozygous for an Mitf null allele, and it induces such a transdifferentiation in embryos that are either heterozygous for the Mitf null allele or homozygous for an RPE-specific hypomorphic Mitf allele generated by targeted mutation. Conversely, an increase in Pax6 gene dose interferes with transdifferentiation even in homozygous Mitf null embryos. Gene expression analyses show that, together with MITF or its paralog TFEC, PAX6 suppresses the expression of Fgf15 and Dkk3. Explant culture experiments indicate that a combination of FGF and DKK3 promote retina formation by inhibiting canonical WNT signaling and stimulating the expression of retinogenic genes, including Six6 and Vsx2. Our results demonstrate that in conjunction with Mitf/Tfec Pax6 acts as an anti-retinogenic factor, whereas in conjunction with retinogenic genes it acts as a pro-retinogenic factor. The results suggest that careful manipulation of the Pax6 regulatory circuit may facilitate the generation of retinal and pigment epithelium cells from embryonic or induced pluripotent stem cells.


Subject(s)
Eye Proteins , Homeodomain Proteins , Microphthalmia-Associated Transcription Factor , Paired Box Transcription Factors , Repressor Proteins , Retina/growth & development , Retinal Pigment Epithelium/growth & development , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing , Animals , Cell Transdifferentiation , Embryonic Development , Eye Proteins/genetics , Eye Proteins/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Dosage , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Wnt Signaling Pathway/genetics
6.
Development ; 138(5): 873-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21247964

ABSTRACT

The pituitary gland is an endocrine organ that is developmentally derived from a fold in the oral ectoderm and a juxtaposed fold in the neural ectoderm. Here, we show that the absence of Vax1, a homeodomain transcription factor known for its role in eye and optic chiasm development, causes the rostral oral ectoderm to form an ectopic fold that eventually develops into a separate second pituitary with all the pituitary cell types and neuronal fibers characteristic of the normal pituitary. The induction of the second pituitary is associated with a localized ectopic expression of Fgf10, a gene encoding a growth factor known to recruit oral ectodermal cells into the pituitary. Interestingly, there are rare cases of pituitary duplications in humans that are also associated with optic nerve dysplasia, suggesting that VAX1 might be involved in the pathogenesis of this disorder.


Subject(s)
Ectoderm/cytology , Neuropeptides/deficiency , Pituitary Gland/growth & development , Transcription Factors/deficiency , Animals , Fibroblast Growth Factor 10/biosynthesis , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Nerve Fibers , Optic Nerve Diseases/etiology , Pituitary Gland/innervation , Transcription Factors/genetics , Transcription Factors/physiology
7.
bioRxiv ; 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37786677

ABSTRACT

MITF, a basic-Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. To explore MITF regulation and its role in melanoma, we conducted a genetic screen for suppressors of the Mitf-associated pigmentation phenotype. An intragenic Mitf mutation was identified, leading to termination of MITF at the K316 SUMOylation site and loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. Interestingly, as a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability and ensuring an active nuclear MITF supply. Interactions between K316 SUMOylation and S409 phosphorylation sites across monomers largely explain the observed effects. Notably, the recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409, unraveling a novel regulatory mechanism with unexpected disease insights.

8.
Dev Biol ; 332(2): 383-95, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19520072

ABSTRACT

Meox1 and Meox2 are two related homeodomain transcription factor genes that together are essential for the development of all somite compartments. Here we show that mice homozygous for Meox1 mutations alone have abnormalities that are restricted to the sclerotome and its derivatives. A prominent and consistent phenotype of these mutations is a remodeling of the cranio-cervical joints whose major feature is the assimilation of the atlas into the basioccipital bone so that the skull rests on the axis. These abnormalities can be traced back to changes in the relative rates of cell proliferation in the rostral and caudal sclerotome compartments, and they are associated with alterations in the expression of at least three transcription factor genes, Tbx18, Uncx, and Bapx1. As previously observed for Bapx1, MEOX1 protein occupies evolutionarily conserved promoter regions of Tbx18 and Uncx, suggesting that Meox1 regulates these genes at least in part directly. Hence, Meox1 is part of a regulatory circuit that serves an essential, non-redundant function in the maintenance of rostro-caudal sclerotome polarity and axial skeleton formation.


Subject(s)
Body Patterning/physiology , Cervical Vertebrae/embryology , Homeodomain Proteins/metabolism , Joints/embryology , Mesoderm/metabolism , Skull/embryology , Animals , Biomarkers/metabolism , Cervical Vertebrae/abnormalities , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , In Situ Hybridization , Joints/abnormalities , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Promoter Regions, Genetic , Skull/abnormalities , Somites/cytology , Somites/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Nat Commun ; 11(1): 333, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949145

ABSTRACT

Cutaneous malignant melanoma is an aggressive cancer of melanocytes with a strong propensity to metastasize. We posit that melanoma cells acquire metastatic capability by adopting an embryonic-like phenotype, and that a lineage approach would uncover metastatic melanoma biology. Using a genetically engineered mouse model to generate a rich melanoblast transcriptome dataset, we identify melanoblast-specific genes whose expression contribute to metastatic competence and derive a 43-gene signature that predicts patient survival. We identify a melanoblast gene, KDELR3, whose loss impairs experimental metastasis. In contrast, KDELR1 deficiency enhances metastasis, providing the first example of different disease etiologies within the KDELR-family of retrograde transporters. We show that KDELR3 regulates the metastasis suppressor, KAI1, and report an interaction with the E3 ubiquitin-protein ligase gp78, a regulator of KAI1 degradation. Our work demonstrates that the melanoblast transcriptome can be mined to uncover targetable pathways for melanoma therapy.


Subject(s)
Gene Expression Profiling , Melanoma/genetics , Melanoma/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcriptome , Animals , Cell Line, Tumor , Endoplasmic Reticulum , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kangai-1 Protein/genetics , Kangai-1 Protein/metabolism , Lung/pathology , Melanocytes/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasm Metastasis/genetics , Neoplasms, Second Primary/pathology , Phenotype , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Skin Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Melanoma, Cutaneous Malignant
10.
Genetics ; 178(1): 259-72, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18202372

ABSTRACT

The mouse Mitf gene encodes a transcription factor that is regulated by serine phosphorylation and is critical for the development of melanin-containing pigment cells. To test the role of phosphorylation at a particular serine, S73 in exon 2 of Mitf, we used a standard targeting strategy in mouse embryonic stem cells to change the corresponding codon into one encoding an alanine. By chance, we generated an allele in which 85,222 bp of wild-type Mitf sequence are duplicated and inserted into an otherwise correctly targeted Mitf gene. Depending on the presence or absence of a neomycin resistance cassette, this genomic rearrangement leads to animals with a white coat with or without pigmented spots or a gray coat with obligatory white and black spots. Several independent, genetically stable germline revertants that lacked the duplicated wild-type sequence but retained the targeted codon were then derived. These animals were normally pigmented, indicating that the serine-to-alanine mutation is not deleterious to melanocyte development. The fact that mosaic coat reversions occur in all mice lacking the neo-cassette and that approximately 1% of these transmit a reverted allele to their offspring places this mutation among those with the highest spontaneous reversion rates in mammals.


Subject(s)
Alleles , Germ-Line Mutation/genetics , Microphthalmia-Associated Transcription Factor/genetics , Alanine/genetics , Amino Acid Substitution/drug effects , Animals , Drug Resistance/drug effects , Female , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Gene Targeting , Homozygote , In Situ Hybridization, Fluorescence , Introns/genetics , Male , Mice , Mutagenesis, Insertional/drug effects , Neomycin/pharmacology , Phenotype , Pigmentation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Serine/genetics
11.
Prog Retin Eye Res ; 73: 100766, 2019 11.
Article in English | MEDLINE | ID: mdl-31242455

ABSTRACT

Dysfunction and loss of the retinal pigment epithelium (RPE) are hallmarks of retinal degenerative diseases in mammals. A critical transcription factor for RPE development and function is the microphthalmia-associated transcription factor MITF and its germline mutations are associated with clinically distinct disorders, including albinism, microphthalmia, retinal degeneration, and increased risk of developing melanoma. Many studies have revealed new insights into central roles of MITF in RPE cell physiology, including melanogenesis, regulation of trophic factor expression, cell proliferation, anti-oxidant functions, and the visual cycle. In this review, we discuss the complex functional roles of MITF in RPE development, homeostasis, and retinal degeneration and touch upon key questions and challenges in neuroprotective strategies for retinal degenerative disorders associated with deficiencies in MITF or its many target genes.


Subject(s)
Microphthalmia-Associated Transcription Factor/physiology , Retinal Degeneration/physiopathology , Retinal Pigment Epithelium/physiology , Animals , Gene Expression Regulation/physiology , Humans
12.
Annu Rev Virol ; 5(1): 33-51, 2018 09 29.
Article in English | MEDLINE | ID: mdl-29958082

ABSTRACT

The discovery of the Mx gene-dependent, innate resistance of mice against influenza virus was a matter of pure chance. Although the subsequent analysis of this antiviral resistance was guided by straightforward logic, it nevertheless led us into many blind alleys and was full of surprising turns and twists. Unexpectedly, this research resulted in the identification of one of the first interferon-stimulated genes and provided a new view of interferon action. It also showed that in many species, MX proteins have activities against a broad range of viruses. To this day, Mx research continues to flourish and to provide insights into the never-ending battle between viruses and their hosts.


Subject(s)
Biomedical Research/history , Disease Resistance , Immunity, Innate , Myxovirus Resistance Proteins/metabolism , Viruses/immunology , Animals , History, 20th Century , History, 21st Century , Humans , Interferons/metabolism
13.
Invest Ophthalmol Vis Sci ; 59(15): 6067-6073, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30590377

ABSTRACT

Purpose: Complete deficiency of microphthalmia transcription factor (MITF) in Mitfmi-vga9/mi-vga9 mice is associated with microphthalmia, retinal dysplasia, and albinism. We investigated the ability of dopachrome tautomerase (DCT) promoter-mediated inducible ectopic expression of Mitf-M to rescue these phenotypic abnormalities. Methods: A new mouse line was created with doxycycline-inducible ectopic Mitf-M expression on an Mitf-deficient Mitfmi-vga9 background (DMV mouse). Adult DMV mice were phenotypically characterized and tissues were collected for histology, immunohistochemistry, and evaluation of Mitf, pigmentary genes, and retinal pigment epithelium (RPE) gene expression. Results: Ectopic Mitf-M expression was specifically induced in the eyes, but was not detected in the skin of DMV mice. Inducible expression of Mitf-M partially rescued the microphthalmia, RPE structure, and pigmentation as well as a subset of the choroidal and iris melanocytes but not cutaneous melanocytes. RPE function and vision were not restored in the DMV mice. Conclusions: Ectopic expression of Mitf-M during development of Mitf-deficient mice is capable of partially rescuing ocular and retinal structures and uveal melanocytes. These findings provide novel information about the roles of Mitf isoforms in the development of mouse eyes.


Subject(s)
Ectopic Gene Expression/physiology , Gene Expression Regulation, Developmental/physiology , Microphthalmia-Associated Transcription Factor/genetics , Retinal Pigment Epithelium/metabolism , Animals , Blotting, Western , Choroid/cytology , Embryonic Development , Female , Gene Expression Profiling , Genotyping Techniques , Immunohistochemistry , Intramolecular Oxidoreductases/pharmacology , Iris/cytology , Male , Melanocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microphthalmos/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/cytology
14.
Pigment Cell Melanoma Res ; 34(1): 7-9, 2021 01.
Article in English | MEDLINE | ID: mdl-33089634
15.
Genetics ; 163(1): 267-76, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12586714

ABSTRACT

Mutations at the mouse microphthalmia locus (Mitf) affect the development of different cell types, including melanocytes, retinal pigment epithelial cells of the eye, and osteoclasts. The MITF protein is a member of the MYC supergene family of basic-helix-loop-helix-leucine-zipper (bHLHZip) transcription factors and is known to regulate the expression of cell-specific target genes by binding DNA as homodimer or as heterodimer with related proteins. The many mutations isolated at the locus have different effects on the phenotype and can be arranged in an allelic series in which the phenotypes range from near normal to white microphthalmic animals with osteopetrosis. Previous investigations have shown that certain combinations of Mitf alleles complement each other, resulting in a phenotype more normal than that of each homozygote alone. Here we analyze this interallelic complementation in detail and show that it is limited to one particular allele, Mitf(Mi-white) (Mitf(Mi-wh)), a mutation affecting the DNA-binding domain. Both loss- and gain-of-function mutations are complemented, as are other Mitf mutations affecting the DNA-binding domain. Furthermore, this behavior is not restricted to particular cell types: Both eye development and coat color phenotypes are complemented. Our analysis suggests that Mitf(Mi-wh)-associated interallelic complementation is due to the unique biochemical nature of this mutation.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Complementation Test , Transcription Factors/genetics , Animals , Eye Abnormalities , Heterozygote , Mice , Microphthalmia-Associated Transcription Factor , Mutation
16.
Genetics ; 167(1): 233-41, 2004 May.
Article in English | MEDLINE | ID: mdl-15166150

ABSTRACT

The MITF protein is a member of the MYC family of basic helix-loop-helix leucine zipper (bHLH-Zip) transcription factors and is most closely related to the TFE3, TFEC, and TFEB proteins. In the mouse, MITF is required for the development of several different cell types, including the retinal pigment epithelial (RPE) cells of the eye. In Mitf mutant mice, the presumptive RPE cells hyperproliferate, abnormally express the retinal transcriptional regulator Pax6, and form an ectopic neural retina. Here we report the structure of the Mitf gene in Drosophila and demonstrate expression during embryonic development and in the eye-antennal imaginal disc. In vitro, transcriptional regulation by Drosophila Mitf, like its mouse counterpart, is modified by the Eyeless (Drosophila Pax6) transcription factor. In vivo, targeted expression of wild-type or dominant-negative Drosophila Mitf results in developmental abnormalities reminiscent of Mitf function in mouse eye development. Our results suggest that the Mitf gene is the original member of the Mitf-Tfe subfamily of bHLH-Zip proteins and that its developmental function is at least partially conserved between vertebrates and invertebrates. These findings further support the common origin of the vertebrate and invertebrate eyes.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Drosophila melanogaster/genetics , Eye/embryology , Gene Expression Regulation, Developmental , Transcription Factors/chemistry , Transcription Factors/physiology , Alleles , Amino Acid Sequence , Animals , Conserved Sequence , DNA, Complementary/metabolism , Evolution, Molecular , Genes, Dominant , Genome , Mice , Microphthalmia-Associated Transcription Factor , Models, Genetic , Molecular Sequence Data , Mutation , Pigment Epithelium of Eye/embryology , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transcription, Genetic , Transcriptional Activation , Transfection
17.
PLoS One ; 8(3): e59247, 2013.
Article in English | MEDLINE | ID: mdl-23555005

ABSTRACT

During vertebrate eye development, the transcription factor MITF acts to promote the development of the retinal pigment epithelium (RPE). In embryos with Mitf mutations, the future RPE hyperproliferates and is respecified as retinal tissue but only in a small portion of the dorsal RPE. Using a series of genetic crosses, we show that this spatial restriction of RPE respecification is brought about by persistent expression of the anti-retinogenic ventral homeodomain gene Vax2 in the dorso-proximal and both Vax1 and Vax2 in the ventral RPE. We further show that dorso-proximal RPE respecification in Vax2/Mitf double mutants and dorso-proximal and ventral RPE respecification in Vax1/2/Mitf triple mutants result from increased FGF/MAP kinase signaling. In none of the mutants, however, does the distal RPE show signs of hyperproliferation or respecification, likely due to local JAGGED1/NOTCH signaling. Expression studies and optic vesicle culture experiments also suggest a role for NOTCH signaling within the mutant dorsal RPE domains, where ectopic JAGGED1 expression may partially counteract the effects of FGF/ERK1/2 signaling on RPE respecification. The results indicate the presence of complex interplays between distinct transcription factors and signaling molecules during eye development and show how RPE phenotypes associated with mutations in one gene are modulated by expression changes in other genes.


Subject(s)
Epistasis, Genetic , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Microphthalmia-Associated Transcription Factor/genetics , Neuropeptides/genetics , Retinal Pigment Epithelium/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Differentiation , Cell Proliferation , Crosses, Genetic , Embryo, Mammalian , Epithelial Cells/cytology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Mutation , Neuropeptides/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/embryology , Serrate-Jagged Proteins , Signal Transduction
18.
Pigment Cell Melanoma Res ; 30(6): 507, 2017 01.
Article in English | MEDLINE | ID: mdl-29094508
19.
Pigment Cell Melanoma Res ; 30(3): 279, 2017 05.
Article in English | MEDLINE | ID: mdl-28460418
20.
Pigment Cell Melanoma Res ; 30(4): 383, 2017 07.
Article in English | MEDLINE | ID: mdl-28636210
SELECTION OF CITATIONS
SEARCH DETAIL