Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neurosci Lett ; 699: 16-23, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30664902

ABSTRACT

Transplantation of dopaminergic precursors (DPs) is a promising therapeutic strategy of Parkinson's disease (PD). However, limited cell source for dopaminergic precursors has become a major obstacle for transplantation therapy. Our group demonstrated previously that mouse fibroblasts can be reprogrammed into induced dopaminergic precursors (iDPs) with high differentiation efficiency. In the current study, we hypothesized that a similar strategy can be applied to generate human iDPs for future cell therapy of PD. We overexpressed transcription factors Brn2, Sox2, and Foxa2 in human fibroblasts and observed formation of neurospheres. Subsequent characterization of the precursor colonies confirmed the generation of human induced dopaminergic precursors (hiDPs). These hiDPs were capable of self-renewal, proliferation, and differentiation. The hiDPs demonstrated high immunoreactivity for neural progenitor markers and high levels of gene expression for ventral mesencephalon-related neural progenitor markers such as Lmx1a, NIKX6.1, Corin, Otx2 and Mash1. Furthermore, the hiDPs could be differentiated into dopaminergic neurons with ˜80% efficiency, which significantly increased major functionally relevant proteins such as TH, DAT, AADC, Lmx1B, and VMAT2 compared to hiDPs. Additionally, hiDPs are more dopaminergic progenitor-restricted compare to those hiDP-like cells reprogrammed only by Brn2 and Sox2. Together, these results suggest that hiDPs with high differentiation efficiency can be generated by direct lineage reprogramming of fibroblasts with transcription factors Brn2, Sox2, and Foxa2. These hiDPs may serve as a safe and effective cell source for transplantation treatment of PD.


Subject(s)
Cell Differentiation , Cell Lineage/physiology , Dopaminergic Neurons/cytology , Fibroblasts/cytology , Biomarkers/metabolism , Cell Proliferation/physiology , Cell Self Renewal/physiology , Cell Transplantation , Cells, Cultured , Genetic Vectors , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , POU Domain Factors/biosynthesis , Retroviridae , SOXB1 Transcription Factors/biosynthesis , Transfection
2.
PLoS Genet ; 10(6): e1004343, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945799

ABSTRACT

Specialized endocrine cells produce and release steroid hormones that govern development, metabolism and reproduction. In order to synthesize steroids, all the genes in the biosynthetic pathway must be coordinately turned on in steroidogenic cells. In Drosophila, the steroid producing endocrine cells are located in the prothoracic gland (PG) that releases the steroid hormone ecdysone. The transcriptional regulatory network that specifies the unique PG specific expression pattern of the ecdysone biosynthetic genes remains unknown. Here, we show that two transcription factors, the POU-domain Ventral veins lacking (Vvl) and the nuclear receptor Knirps (Kni), have essential roles in the PG during larval development. Vvl is highly expressed in the PG during embryogenesis and is enriched in the gland during larval development, suggesting that Vvl might function as a master transcriptional regulator in this tissue. Vvl and Kni bind to PG specific cis-regulatory elements that are required for expression of the ecdysone biosynthetic genes. Knock down of either vvl or kni in the PG results in a larval developmental arrest due to failure in ecdysone production. Furthermore, Vvl and Kni are also required for maintenance of TOR/S6K and prothoracicotropic hormone (PTTH) signaling in the PG, two major pathways that control ecdysone biosynthesis and PG cell growth. We also show that the transcriptional regulator, Molting defective (Mld), controls early biosynthetic pathway steps. Our data show that Vvl and Kni directly regulate ecdysone biosynthesis by transcriptional control of biosynthetic gene expression and indirectly by affecting PTTH and TOR/S6K signaling. This provides new insight into the regulatory network of transcription factors involved in the coordinated regulation of steroidogenic cell specific transcription, and identifies a new function of Vvl and Knirps in endocrine cells during post-embryonic development.


Subject(s)
Drosophila Proteins/metabolism , Ecdysone/biosynthesis , Insect Hormones/biosynthesis , Nuclear Proteins/metabolism , POU Domain Factors/metabolism , Repressor Proteins/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Animals , Binding Sites , Biological Transport/genetics , Cholesterol/metabolism , DNA-Binding Proteins , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Ecdysone/genetics , Ecdysone/metabolism , Gene Expression Regulation , Gene Expression Regulation, Developmental , Insect Hormones/metabolism , Membrane Proteins/biosynthesis , POU Domain Factors/biosynthesis , POU Domain Factors/genetics , RNA Interference , RNA, Small Interfering , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , TOR Serine-Threonine Kinases/biosynthesis , Transcription, Genetic
3.
Pigment Cell Melanoma Res ; 27(3): 442-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24472175

ABSTRACT

Spontaneous melanoma phenotype switching is controlled by unknown environmental factors and may determine melanoma outcome and responsiveness to anticancer therapy. We show that Orai1 and STIM2 are highly expressed and control store-operated Ca(2+) entry in human melanoma. Lower extracellular Ca(2+) or silencing of Orai1/STIM2 caused a decrease in intracellular Ca(2+) , which correlated with enhanced proliferation and increased expression of microphthalmia-associated transcription factor, a marker for proliferative melanoma phenotype. In contrast, the invasive and migratory potential of melanoma cells was reduced upon silencing of Orai1 and/or STIM2. Accordingly, markers for a non-proliferative, tumor-maintaining phenotype such as JARID1B and Brn2 decreased. Immunohistochemical staining of primary melanomas and lymph node metastases revealed a heterogeneous distribution of Orai1 and STIM2 with elevated expression in the invasive rim of the tumor. In summary, our results support a dynamic model in which Orai1 and STIM2 inversely control melanoma growth and invasion. Pharmacological tuning of Orai1 and particularly STIM2 might thus prevent metastatic spread and render melanomas more susceptible to conventional therapy.


Subject(s)
Calcium Channels/physiology , Calcium Signaling/physiology , Cell Adhesion Molecules/physiology , Melanoma/pathology , Neoplasm Proteins/physiology , Animals , Brain Neoplasms/secondary , Calcium Channel Blockers/pharmacology , Calcium Channels/genetics , Cattle , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Division , Cell Line, Tumor , Cell Movement , Culture Media/pharmacology , Extracellular Fluid/metabolism , Glycosylation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Ion Transport/drug effects , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Jumonji Domain-Containing Histone Demethylases/genetics , Lymphatic Metastasis , Melanins/biosynthesis , Melanoma/metabolism , Melanoma/secondary , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , ORAI1 Protein , POU Domain Factors/biosynthesis , POU Domain Factors/genetics , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Serum , Stromal Interaction Molecule 2
4.
Int J Dev Neurosci ; 31(1): 82-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085081

ABSTRACT

Neural stem cells (NSCs) tranplantation has great potential for the treatment of neurodegenerative disease such as Parkinson's disease (PD). However, the usage of NSCs is limited because the differentiation of NSCs into specific dopaminergic neurons has proven difficult. We have recently demonstrated that transgenic expression of Nurr1 could induce the differentiation of NSCs into tyrosine hydroxylase (TH) immunoreactive dopaminergic neurons, and forced co-expression of Nurr1 with Brn4 caused a dramatic increase in morphological and phenotypical maturity of these neurons. In this study, we investigated the effect of transplanted NSCs in PD model rats. The results showed that overexpression of Nurr1 promoted NSCs to differentiate into dopaminergic neurons in vivo, increased the level of dopamine (DA) neurotransmitter in the striatum, resulting in behavioral improvement of PD rats. Importantly, co-expression of Nurr1 and Brn4 in NSCs significantly increased the maturity and viability of dopaminergic neurons, further raised the DA amount in the striatum and reversed the behavioral deficit of the PD rats. Our findings provide a new potential and strategy for the use of NSCs in cell replacement therapy for PD.


Subject(s)
Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , POU Domain Factors/metabolism , Parkinsonian Disorders/surgery , Stem Cell Transplantation/methods , Adrenergic Agents/toxicity , Animals , Apomorphine , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Agonists , Dopamine Plasma Membrane Transport Proteins/metabolism , Green Fluorescent Proteins/genetics , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Oxidopamine/toxicity , POU Domain Factors/biosynthesis , POU Domain Factors/genetics , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Transfection , Tyrosine 3-Monooxygenase/metabolism
5.
Dev Growth Differ ; 54(7): 702-16, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22957893

ABSTRACT

POU-V class proteins like Oct4 are crucial for keeping cells in an undifferentiated state. An Oct4 homologue in Xenopus laevis, Oct25, peaks in expression during early gastrulation, when many cells are still uncommitted. Nevertheless, extensive morphogenesis is taking place in all germ layers at that time. Phenotypical analysis of embryos with Oct25 overexpression revealed morphogenesis defects, beginning during early gastrulation and resulting in spina-bifida-like axial defects. Analysis of marker genes and different morphogenesis assays show inhibitory effects on convergence and extension and on mesoderm internalization. On a cellular level, cell-cell adhesion is reduced. On a molecular level, Oct25 overexpression activates expression of PAPC, a functional inhibitor of the cell adhesion molecule EP/C-cadherin. Intriguingly, Oct25 effects on cell-cell adhesion can be restored by overexpression of EP/C-cadherin or by inhibition of the PAPC function. Thus, Oct25 affects morphogenesis via activation of PAPC expression and subsequent functional inhibition of EP/C-cadherin.


Subject(s)
Cadherins/biosynthesis , Gastrulation/physiology , Gene Expression Regulation, Developmental/physiology , Mesoderm/embryology , Morphogenesis/physiology , POU Domain Factors/biosynthesis , Xenopus Proteins/biosynthesis , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/physiology , Mesoderm/cytology , POU Domain Factors/genetics , Protocadherins , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
6.
PLoS One ; 7(7): e41506, 2012.
Article in English | MEDLINE | ID: mdl-22859994

ABSTRACT

Recent findings have demonstrated that the overexpression of lineage-specific transcription factors induces cell fate changes among diverse cell types. For example, neurons can be generated from mouse and human fibroblasts. It is well known that neurons are terminally differentiated cells that do not divide. Therefore, we consider how to induce glioma cells to become neurons by introducing transcription factors. Here, we describe the efficient generation of induced neuronal (iN) cells from glioma cells by the infection with three transcription factors: Ascl1, Brn2 and Ngn2 (ABN). iN cells expressed multiple neuronal markers and fired action potentials, similar to the properties of authentic neurons. Importantly, the proliferation of glioma cells following ABN overexpression was dramatically inhibited in both in vitro and in vivo experiments. In addition, iN cells that originated from human glioma cells did not continue to grow when they were sorted and cultured in vitro. The strategies by which glioma cells are induced to become neurons may be used to clinically study methods for inhibiting tumor growth.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Transformation, Neoplastic , Homeodomain Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , POU Domain Factors/physiology , Action Potentials , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , G1 Phase Cell Cycle Checkpoints , Glioma , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , POU Domain Factors/biosynthesis , POU Domain Factors/genetics , Transduction, Genetic
7.
BMC Dev Biol ; 11: 52, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21880149

ABSTRACT

BACKGROUND: Nkx2.2 and Arx represent key transcription factors implicated in the specification of islet cell subtypes during pancreas development. Mice deficient for Arx do not develop any alpha-cells whereas beta- and delta-cells are found in considerably higher numbers. In Nkx2.2 mutant animals, alpha- and beta-cell development is severely impaired whereas a ghrelin-expressing cell population is found augmented.Notably, Arx transcription is clearly enhanced in Nkx2.2-deficient pancreata. Hence in order to precise the functional link between both factors we performed a comparative analysis of Nkx2.2/Arx single- and double-mutants but also of Pax6-deficient animals. RESULTS: We show that most of the ghrelin+ cells emerging in pancreata of Nkx2.2- and Pax6-deficient mice, express the alpha-cell specifier Arx, but also additional beta-cell related genes. In Nkx2.2-deficient mice, Arx directly co-localizes with iAPP, PC1/3 and Pdx1 suggesting an Nkx2.2-dependent control of Arx in committed beta-cells. The combined loss of Nkx2.2 and Arx likewise results in the formation of a hyperplastic ghrelin+ cell population at the expense of mature alpha- and beta-cells. Surprisingly, such Nkx2.2-/-Arx- ghrelin+ cells also express the somatostatin hormone. CONCLUSIONS: Our data indicate that Nkx2.2 acts by reinforcing the transcriptional networks initiated by Pax4 and Arx in early committed beta- and alpha-cell, respectively. Our analysis also suggests that one of the coupled functions of Nkx2.2 and Pax4 is to counteract Arx gene activity in early committed beta-cells.


Subject(s)
Cell Lineage , Ghrelin/biosynthesis , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/biosynthesis , Insulin-Secreting Cells/metabolism , Somatostatin/biosynthesis , Transcription Factors/biosynthesis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Eye Proteins/biosynthesis , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Ghrelin/genetics , Glucagon-Secreting Cells/cytology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Islet Amyloid Polypeptide/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , PAX6 Transcription Factor , POU Domain Factors/biosynthesis , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/deficiency , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/deficiency , Repressor Proteins/genetics , Somatostatin/genetics , Trans-Activators/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Zebrafish Proteins
8.
Int J Neurosci ; 120(3): 192-200, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20374086

ABSTRACT

Neurogenesis in the hippocampus continues throughout adult life and can be regulated by the local microenvironment. To determine whether denervation stimulates neurogenesis in hippocampus, proliferation, migration, and differentiation of local neural stem cells (NSCs) in dentate gyrus was investigated after fimbria fornix transection. In the denervated hippocampus, NSCs proliferated markedly and migrated along the subgranular layer, and more newborn cells differentiated into neurons or astrocytes. After denervation, more newborn cells in the deafferented hippocampus expressed Brn-4 and differentiated into beta-Tubulin III positive neurons. It is concluded that the local NSCs in hippocampus may proliferate and migrate into granule cell layer, in which changes in the deafferented hippocampus provided a suitable microenvironment for hippocampal neurogenesis and the increased Brn-4 in denervated hippocampus may be involved in this process.


Subject(s)
Adult Stem Cells/cytology , Cell Movement , Cell Proliferation , Fornix, Brain/injuries , Hippocampus/cytology , Neurogenesis , Adult Stem Cells/metabolism , Animals , Astrocytes/cytology , Female , Hippocampus/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/cytology , POU Domain Factors/biosynthesis , Rats , Rats, Sprague-Dawley , Staining and Labeling , Tubulin/biosynthesis
9.
Neurosci Res ; 67(1): 8-17, 2010 May.
Article in English | MEDLINE | ID: mdl-20105446

ABSTRACT

Brn-4, a member of the homeobox family of transcription factors, has previously been implicated in the regeneration and repair of denervated striatum. We investigated the effects of Brn-4 on the differentiation and development of neural stem cells (NSCs) from E16 rat hippocampus. Immunocytochemistry revealed that extracts of deafferented hippocampus promoted neuronal differentiation to a greater extent than extracts from normal hippocampus. Deafferented extracts also promoted maturation of newborn neurons as reflected in changes in cell areas and perimeters, and enhanced Brn-4 expression in MAP-2 positive neurons. Suppression or overexpression of Brn-4 in NSCs markedly decreased or increased neuronal differentiation and maturation of newborn neurons, respectively. These results suggest that Brn-4 expression is required both for neuronal differentiation of NSCs and maturation of newborn neurons, and that there may be some regulatory factors in deafferented hippocampus that can regulate Brn-4 expression in neuronal progenitors. Brn-4 is therefore a potential research target for the development of new therapeutics to promote brain repair.


Subject(s)
Nerve Tissue Proteins/physiology , Neurons/cytology , POU Domain Factors/physiology , Stem Cells/cytology , Afferent Pathways , Animals , Cell Differentiation , Embryo, Mammalian , Female , Hippocampus/cytology , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , POU Domain Factors/biosynthesis , RNA Interference , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism , Tissue Extracts/pharmacology
10.
J Neurosci ; 28(31): 7748-64, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18667607

ABSTRACT

The diversity of cell types found within the vertebrate CNS arises in part from action of complex transcriptional programs. In the retina, the programs driving diversification of various cell types have not been completely elucidated. To investigate gene regulatory networks that underlie formation and function of one retinal circuit component, the bipolar cell, transcriptional regulation of three bipolar cell-enriched genes was analyzed. Using in vivo retinal DNA transfection and reporter gene constructs, a 200 bp Grm6 enhancer sequence, a 445 bp Cabp5 promoter sequence, and a 164 bp Chx10 enhancer sequence, were defined, each driving reporter expression specifically in distinct but overlapping bipolar cell subtypes. Bioinformatic analysis of sequences revealed the presence of potential paired-type and POU homeodomain-containing transcription factor binding sites, which were shown to be critical for reporter expression through deletion studies. The paired-type homeodomain transcription factors (TFs) Crx and Otx2 and the POU homeodomain factor Brn2 are expressed in bipolar cells and interacted with the predicted binding sequences as assessed by electrophoretic mobility shift assay. Grm6, Cabp5, and Chx10 reporter activity was reduced in Otx2 loss-of-function retinas. Endogenous gene expression of bipolar cell molecular markers was also dependent on paired-type homeodomain-containing TFs, as assessed by RNA in situ hybridization and reverse transcription-PCR in mutant retinas. Cabp5 and Chx10 reporter expression was reduced in dominant-negative Brn2-transfected retinas. The paired-type and POU homeodomain-containing TFs Otx2 and Brn2 together appear to play a common role in regulating gene expression in retinal bipolar cells.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , POU Domain Factors/physiology , Retinal Bipolar Cells/physiology , Transcription Factors/physiology , Animals , Animals, Newborn , Female , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Mice , Mice, Knockout , Mice, Transgenic , Otx Transcription Factors/biosynthesis , Otx Transcription Factors/deficiency , Otx Transcription Factors/genetics , POU Domain Factors/biosynthesis , POU Domain Factors/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Trans-Activators/biosynthesis , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
11.
Biochem Biophys Res Commun ; 353(3): 644-9, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17194450

ABSTRACT

We have previously indicated that Xenopus Suppressor of Hairless 2, XSu(H)2, is involved in the morphogenesis of gastrula embryos in a different manner from the XESR-1-mediated Notch signaling pathway. To address the downstream factors of XSu(H)2, we investigated the effect of XSu(H)2 on XenopusPOU-V genes, Xoct25 and Xoct91. Knockdown of XSu(H)2 caused the downregulation of Xoct25, Xoct91 and Xbrachyury. Dominant-negative Xoct25 caused the delay of blastopore closure with a decrease of Xbrachyury expression. Overexpression of Xoct25 or Xoct91 could rescue the decrease of Xbrachyury expression caused by XSu(H)2 depletion. XSu(H)2EnR inhibited Xoct25 and Xoct91 expressions in CHX-treated animal caps. Promoter analysis of the Xoct25 showed that the upstream region of Xoct25 contains the prospective Su(H) binding motif, which is essential for the transcription of Xoct25. These results suggest that XSu(H)2 is engaged in the cell fate decision during gastrulation through the gene expression of the Xoct25/91-mediated pathway.


Subject(s)
Gastrula/physiology , Gene Expression Regulation, Developmental , Octamer Transcription Factors/physiology , Transcription Factors/physiology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Animals , POU Domain Factors/biosynthesis
12.
J Pediatr Hematol Oncol ; 28(12): 791-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17164647

ABSTRACT

We have previously suggested the transcription factor gene POU6F2 as a novel tumor suppressor involved in Wilms tumor (WT) predisposition. Since WT arises from pluripotent embryonic renal precursors, in this study we analyzed the expression of the murine homolog Pou6f2 during kidney embryogenesis and compared it to that of Wt1, the homolog of WT1, a known WT related gene involved in mesenchyme to epithelium conversion. Quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) performed for Pou6f2 on kidney specimens from embryos, pups, and adult mice, showed that the Pou6f2 mRNA was more abundant in the earliest analyzed phase of kidney organogenesis (E13) than in more advanced fetal stages and in adult animal. In situ RT-PCR demonstrated that Pou6f2 expression parallels the centripetal differentiation of renal morphogenesis. In addition, in E18 kidney, most structures exhibiting Pou6f2 expression stained positively in immunohistochemistry for the Wt1 protein. Finally, quantitative real-time RT-PCR revealed an overexpression (>/=80 times) of POU6F2 compared with normal kidney in 5 of 22 (23%) WTs. The finding of a highly regulated temporal and spatial Pou6f2 expression during renal organogenesis, of its coexpression with Wt1 and of POU6F2 overexpression in a subset of WTs are consistent with a role of POU6F2 in kidney development and provide further support to its involvement in WT.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Kidney/embryology , POU Domain Factors/biosynthesis , WT1 Proteins/biosynthesis , Wilms Tumor/metabolism , Adult , Aged , Animals , Cell Differentiation/genetics , Female , Genetic Predisposition to Disease , Humans , Kidney/metabolism , Kidney/pathology , Male , Mesoderm/metabolism , Mesoderm/pathology , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organogenesis/genetics , POU Domain Factors/genetics , WT1 Proteins/genetics , Wilms Tumor/genetics , Wilms Tumor/pathology
13.
Dev Biol ; 286(2): 618-28, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16168406

ABSTRACT

How a common neurotransmitter phenotype specified in neurons of different origins is an outstanding issue in neuronal development and function. In C. elegans larvae, serotonin is synthesized in 2 pairs of neurons, the secretory neurons NSM and the chemosensory neurons ADF. In order to delineate the molecular mechanisms of serotonergic phenotype establishment, we have screened for neuron-specific serotonin deficient (nss) mutants. Our prior study showed that the POU-homeodomain factor UNC-86 is expressed in and required for the NSM neurons to adopt serotonergic phenotype and correct pathfinding, whereas ADF are unaffected in unc-86-null mutants. Here, we report that the LIM-homeodomain factor LIM-4 regulates ADF serotonergic phenotype. In lim-4 mutants, many aspects of ADF differentiation occur, however, they fail to express serotonin phenotype and exhibit aberrant cilia properties. LIM-4 expression rises in the neuroblast that produces two distinct neurons: ADF and the olfactory neuron AWB. We show that lim-4 is regulated by separable mechanisms to determine disparate subtype identities in these two neuronal types. In vivo promoter analyses reveal that cis-element(s) within introns are necessary and sufficient to direct lim-4 to specify serotonergic phenotype, whereas its 5'-upstream sequence directs lim-4 function in AWB. Thus, a transcription factor may act independently to specify distinct differentiation traits in two sister cells. We propose that serotonergic identity is specified in cell-specific contexts to coordinate the development and function.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Homeodomain Proteins/physiology , Neurons/metabolism , Recombinant Fusion Proteins/physiology , Serotonin/biosynthesis , Transcription Factors/physiology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/genetics , Enhancer Elements, Genetic , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Mutation , Neurons/cytology , POU Domain Factors/biosynthesis , POU Domain Factors/physiology , Phenotype , Recombinant Fusion Proteins/genetics , Serotonin/deficiency , Transcription Factors/genetics
15.
Dev Biol ; 281(2): 286-98, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15893979

ABSTRACT

A specific prediction of our detailed cis-regulatory analysis of the Strongylocentrotus purpuratus (Sp) endo16 gene was that the later expression of this gene would be driven by a midgut-specific transcriptional regulator. We have now identified this factor and determined some of its functions. The cDNA sequence reveals it to be a POU domain factor related closely to the mammalian factors Brain-1, -2, and -4. The factor was termed SpBrn1/2/4 (henceforth Brn1/2/4). Quantitative measurements of transcript prevalence show that the gene is first activated in the 20-h blastula, but there remain only about 100 molecules of brn1/2/4 mRNA per embryo (only a few per endoderm cell) until an abrupt 10-fold increase occurs as gastrulation begins. Measured in the same embryos, the late rise in prevalence of endo16 transcripts follows that of brn1/2/4 transcripts. As predicted by the endo16 model, brn1/2/4 expression is confined perfectly to the midgut, coincident with the domain of endo16 expression. The kinetics of accumulation of these transcripts indicates that the switch into the late phase of endo16 expression occurs when the brn1/2/4 transcript level nears its plateau (2000 molecules mRNA per embryo), after which each endo16 gene produces about 1 mRNA every 2 min (about 380 molecules mRNA per min in the whole embryo). Arrest of Brn1/2/4 translation by MASO treatment blocks the late phase of endo16 expression and specifically abolishes expression of cis-regulatory Module B of endo16, while not affecting Module A, also as predicted. The brn1/2/4 gene lies downstream of the regulatory genes executing post-gastrular specification of the midgut, as shown by further gene expression perturbation experiments which provide an initial glimpse of the underlying network architecture.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Endoderm/physiology , Homeodomain Proteins/physiology , POU Domain Factors/physiology , Sea Urchins/physiology , Amino Acid Sequence , Animals , Cell Adhesion Molecules/genetics , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Molecular Sequence Data , POU Domain Factors/biosynthesis , Proteins/genetics , RNA, Messenger/metabolism , Sea Urchins/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...