Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
G3 (Bethesda) ; 11(8)2021 08 07.
Article in English | MEDLINE | ID: mdl-34849808

ABSTRACT

Male reproduction encompasses many essential cellular processes and interactions. As a focal point for these events, sperm offer opportunities for advancing our understanding of sexual reproduction at multiple levels during development. Using male sterility genes identified in human, mouse, and fruit fly databases as a starting point, 103 Drosophila melanogaster genes were screened for their association with male sterility by tissue-specific RNAi knockdown and CRISPR/Cas9-mediated mutagenesis. This list included 56 genes associated with male infertility in the human databases, but not found in the Drosophila database, resulting in the discovery of 63 new genes associated with male fertility in Drosophila. The phenotypes identified were categorized into six distinct classes affecting sperm development. Interestingly, the second largest class (Class VI) caused sterility despite apparently normal testis and sperm morphology suggesting that these proteins may have functions in the mature sperm following spermatogenesis. We focused on one such gene, Rack 1, and found that it plays an important role in two developmental periods, in early germline cells or germline stem cells and in spermatogenic cells or sperm. Taken together, many genes are yet to be identified and their role in male reproduction, especially after ejaculation, remains to be elucidated in Drosophila, where a wealth of data from human and other model organisms would be useful.


Subject(s)
Drosophila Proteins , Infertility, Male , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Infertility, Male/genetics , Male , Spermatogenesis/genetics , Testis
2.
Sci Rep ; 11(1): 19991, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620982

ABSTRACT

Poly(ethylene terephthalate) (PET) is a widely used plastic in bottles and fibers; its waste products pollute the environment owing to its remarkable durability. Recently, Ideonella sakaiensis 201-F6 was isolated as a unique bacterium that can degrade and assimilate PET, thus paving the way for the bioremediation and bioconversion of PET waste. We found that this strain harbors a poly(hydroxyalkanoate) (PHA) synthesis gene cluster, which is highly homologous with that of Cupriavidus necator, an efficient PHA producer. Cells grown on PET accumulated intracellular PHA at high levels. Collectively, our findings in this study demonstrate that I. sakaiensis can mediate the direct conversion of non-biodegradable PET into environment-friendly plastic, providing a new approach for PET recycling.

3.
Genes Genet Syst ; 96(4): 177-186, 2021 Dec 16.
Article in English | MEDLINE | ID: mdl-34556622

ABSTRACT

Sperm are modified substantially in passing through both the male and the female reproductive tracts, only thereafter becoming functionally competent to fertilize eggs. Drosophila sperm become motile in the seminal vesicle; after ejaculation, they interact with seminal fluid proteins and undergo biochemical changes on their surface while they are stored in the female sperm storage organs. However, the molecular mechanisms underlying these maturation processes remain largely unknown. Here, we focused on Drosophila Neprilysin genes, which are the fly orthologs of the mouse Membrane metallo-endopeptidase-like 1 (Mmel1) gene. While Mmel1 knockout male mice have reduced fertility without abnormality in either testis morphology or sperm motility, there are inconsistent results regarding the association of any Neprilysin gene with male fertility in Drosophila. We examined the association of the Nep1-5 genes with male fertility by RNAi and found that Nep4 gene function is specifically required in germline cells. To investigate this in more detail, we induced mutations in the Nep4 gene by the CRISPR/Cas9 system and isolated two mutants, both of which were viable and female fertile, but male sterile. The mutant males had normal-looking testes and sperm; during copulation, sperm were transferred to females and stored in the seminal receptacle and paired spermathecae. However, following sperm transfer and storage, three defects were observed for Nep4 mutant sperm. First, sperm were quickly discarded by the females; second, the proportion of eggs fertilized was significantly lower for mutant sperm than for control sperm; and third, most eggs laid did not initiate development after sperm entry. Taking these observations together, we conclude that the Nep4 gene is essential for sperm function following sperm transfer to females.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Fertility/genetics , Male , Mice , Neprilysin/genetics , Sperm Motility/genetics , Spermatozoa
4.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34072894

ABSTRACT

MicroRNA-124 (miR-124) is strongly expressed in neurons, and its expression increases as neurons mature. Through DNA methylation in the miR-124 promoter region and adsorption of miR-124 by non-coding RNAs, miR-124 expression is known to be reduced in many cancer cells, especially with high malignancy. Recently, numerous studies have focused on miR-124 due to its promising tumor-suppressive effects; however, the overview of their results is unclear. We surveyed the tumor-suppressive effect of miR-124 in glial cell lineage cancers, which are the most frequently reported cancer types involving miR-124, and in lung, colon, liver, stomach, and breast cancers, which are the top five causes of cancer death. Reportedly, miR-124 not only inhibits proliferation and accelerates apoptosis, but also comprehensively suppresses tumor malignant transformation. Moreover, we found that miR-124 exerts its anti-tumor effects by regulating a wide range of target genes, most notably STAT3 and EZH2. In addition, when compared to the original role of miR-124 in neuronal development, we found that the miR-124 target genes that contribute to neuronal maturation share similarities with genes that cause cancer cell metastasis and epithelial-mesenchymal transition. We believe that the two apparently unrelated fields, cancer and neuronal development, can bring new discoveries to each other through the study of miR-124.


Subject(s)
Brain/growth & development , MicroRNAs/genetics , Neoplasms/genetics , Neurons/metabolism , Brain/metabolism , Cell Proliferation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasms/pathology , Neuroglia/metabolism , Neuroglia/pathology , STAT3 Transcription Factor/genetics
5.
Am J Med Genet A ; 185(7): 2084-2093, 2021 07.
Article in English | MEDLINE | ID: mdl-33973697

ABSTRACT

Nuclear factor I A (NFIA) is a transcription factor that belongs to the NFI family. Truncating variants or intragenic deletion of the NFIA gene are known to cause the human neurodevelopmental disorder known as NFIA-related disorder, but no patient heterozygous for a missense mutation has been reported. Here, we document two unrelated patients with typical phenotypic features of the NFIA-related disorder who shared a missense variant p.Lys125Glu (K125E) in the NFIA gene. Patient 1 was a 6-year-old female with global developmental delay, corpus callosum anomaly, macrocephaly, and dysmorphic facial features. Patient 2 was a 14-month-old male with corpus callosum anomaly and macrocephaly. By using Drosophila and zebrafish models, we functionally evaluated the effect of the K125E substitution. Ectopic expression of wild-type human NFIA in Drosophila caused developmental defects such as eye malformation and premature death, while that of human NFIA K125E variant allele did not. nfia-deficient zebrafish embryos showed defects of midline-crossing axons in the midbrain/hindbrain boundary. This impairment of commissural neurons was rescued by expression of wild-type human NFIA, but not by that of mutant variant harboring K125E substitution. In accordance with these in vivo functional analyses, we showed that the K125E mutation impaired the transcriptional regulation of HES1 promoter in cultured cells. Taken together, we concluded that the K125E variant in the NFIA gene is a loss-of-function mutation.


Subject(s)
Genetic Predisposition to Disease , Megalencephaly/genetics , NFI Transcription Factors/genetics , Neurodevelopmental Disorders/genetics , Alleles , Amino Acid Substitution/genetics , Animals , Child , Corpus Callosum/metabolism , Corpus Callosum/pathology , Disease Models, Animal , Drosophila/genetics , Female , Gene Expression Regulation, Developmental/genetics , Humans , Infant , Male , Megalencephaly/pathology , Mutation, Missense/genetics , Neurodevelopmental Disorders/pathology , Zebrafish/genetics
6.
Genes Cells ; 25(11): 741-752, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32979863

ABSTRACT

MicroRNAs (miRNAs) are known to regulate gene expression and modulate cellular differentiation. MicroRNA-9/9* (miR-9/9*) and microRNA-124 (miR-124) are highly expressed in the central nervous system. In vivo function of miR-9/9* and miR-124 has been investigated in detail, whereas there remain some discrepancies regarding neural development. To this end, we electroporated miR-9/9*, miR-124 or miR-9/9*/124 expression plasmids into neonatal retinal progenitor cells (RPCs) in vivo and analyzed the fate of electroporated cells. Both miR-9/9* and miR-124 reduced the number of SOX9- and GS-positive cells and increased that of TUBB3-positive cells in the postnatal day 14 retina. No major effects on the proliferation and apoptosis of the electroporated cells were detected at least postnatal day 3. These indicated that miR-9/9* and miR-124 influence the cell fate of glial cells, thereby inducing their differentiation into neurons. Moreover, we found this cell fate modulation was occurred in RPCs indicating high-level expression of miRNA, but not in the low level. Our results strongly suggest that high-level miRNA overexpression is essential for directing cell fate by miR-9/9* and miR-124 interference.


Subject(s)
Ependymoglial Cells/metabolism , MicroRNAs/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/physiology , Ependymoglial Cells/physiology , Female , Gene Expression/physiology , Mice , Mice, Inbred ICR , MicroRNAs/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , Retina/metabolism
7.
Front Biosci (Landmark Ed) ; 25(1): 168-178, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31585884

ABSTRACT

Traumatic brain injury (TBI) causes substantial mortality and disability, but effective treatments are unavailable. An external force causes primary injury, which is followed by secondary injury that triggers chronic neurodegenerative diseases. Therefore, understanding the mechanisms underlying post-TBI secondary injury might provide insights into neurodegenerative diseases. The secondary injury is known to share some physiological features with neurodegenerative diseases. So far, many TBI models in mammals exist, but models in other species are required from the viewpoint of lifespan and animal welfare. In Drosophila, closed and open TBI models are available. Both models have focused on TBI-induced changes in innate immunity. Aging strongly induces innate immunity responses, and neuroinflammation plays an important role in both mammalian models of TBI and humans with TBI. Although Drosophila models do not mimic all phenomena involved in post-TBI secondary injury in mammals, further experiments with Drosophila models and other animal models could elucidate the mechanisms involved in post-TBI secondary brain injury, which would in turn elucidate neurodegenerative processes.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Brain/blood supply , Disease Models, Animal , Drosophila/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Brain/drug effects , Brain/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/prevention & control , Drosophila/immunology , Humans , Longevity/drug effects , Longevity/immunology , Minocycline/administration & dosage , Neuroprotective Agents/administration & dosage
8.
Brain Behav Immun ; 80: 427-438, 2019 08.
Article in English | MEDLINE | ID: mdl-30986429

ABSTRACT

Symptoms of many neurodegenerative diseases appear later in human life. However, young animal models for penetrating traumatic brain injury (pTBI) have been used to study neurodegenerative diseases and evaluate the efficacy of neuroprotective medicines. Possibly because of this discordance, effective neuroprotective drugs have still not been developed. For patients suffering from pTBI, aging is known to be a significant prognostic factor of mortality. In this study, we aimed to establish a model of aged pTBI animals using Drosophila melanogaster. We successfully generated aged pTBI flies as a new pTBI model showing increased neurodegeneration and higher mortality. To elucidate the mechanism of increased vulnerability in aged pTBI animals, we analyzed the GenBank-deposited transcriptome data of young and aged flies, demonstrating the importance of innate immunity genes for higher mortality in aged pTBI models. We found that in the context of pTBI, normal aging strongly activated the expression of antimicrobial peptide genes and upregulated the nuclear factor-κB gene in the immune deficiency pathway, but not the Toll pathway. Moreover, we found that minocycline increased the survival of young pTBI flies, but not aged pTBI flies. These results suggested that immune system activation under neurodegenerative conditions was involved in normal aging, thereby inhibiting the medicinal efficacy of neuroprotective drugs effective for young flies in aged flies.


Subject(s)
Aging/immunology , Brain Injuries/drug therapy , Minocycline/pharmacology , Animals , Brain/metabolism , Brain Injuries/metabolism , Brain Injuries, Traumatic/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Immunity, Innate/immunology , Immunity, Innate/physiology , Male , Models, Animal , Neurodegenerative Diseases/immunology , Neuroprotective Agents/metabolism
9.
Nat Immunol ; 18(12): 1342-1352, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29058703

ABSTRACT

T cells reorganize their metabolic profiles after being activated, but the systemic metabolic effect of sustained activation of the immune system has remained unexplored. Here we report that augmented T cell responses in Pdcd1-/- mice, which lack the inhibitory receptor PD-1, induced a metabolic serum signature characterized by depletion of amino acids. We found that the depletion of amino acids in serum was due to the accumulation of amino acids in activated Pdcd1-/- T cells in the lymph nodes. A systemic decrease in tryptophan and tyrosine led to substantial deficiency in the neurotransmitters serotonin and dopamine in the brain, which resulted in behavioral changes dominated by anxiety-like behavior and exacerbated fear responses. Together these data indicate that excessive activation of T cells causes a systemic metabolomic shift with consequences that extend beyond the immune system.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Fear/physiology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/immunology , Amino Acids/blood , Animals , Brain/metabolism , Dopamine/deficiency , Interferon-gamma/blood , Kynurenine/blood , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Serotonin/deficiency , T-Lymphocytes/metabolism , Tryptophan/metabolism , Tyrosine/metabolism
10.
Mol Cell Biol ; 35(15): 2583-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986607

ABSTRACT

The Rax homeobox gene plays essential roles in multiple processes of vertebrate retina development. Many vertebrate species possess Rax and Rax2 genes, and different functions have been suggested. In contrast, mice contain a single Rax gene, and its functional roles in late retinal development are still unclear. To clarify mouse Rax function in postnatal photoreceptor development and maintenance, we generated conditional knockout mice in which Rax in maturing or mature photoreceptor cells was inactivated by tamoxifen treatment (Rax iCKO mice). When Rax was inactivated in postnatal Rax iCKO mice, developing photoreceptor cells showed a significant decrease in the level of the expression of rod and cone photoreceptor genes and mature adult photoreceptors exhibited a specific decrease in cone cell numbers. In luciferase assays, we found that Rax and Crx cooperatively transactivate Rhodopsin and cone opsin promoters and that an optimum Rax expression level to transactivate photoreceptor gene expression exists. Furthermore, Rax and Crx colocalized in maturing photoreceptor cells, and their coimmunoprecipitation was observed in cultured cells. Taken together, these results suggest that Rax plays essential roles in the maturation of both cones and rods and in the survival of cones by regulating photoreceptor gene expression with Crx in the postnatal mouse retina.


Subject(s)
Eye Proteins/genetics , Homeodomain Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , 3T3 Cells , Animals , Cell Line , Cell Survival , Cone Opsins/biosynthesis , Cone Opsins/genetics , Eye Proteins/metabolism , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Knockout , Neuroglia/metabolism , Promoter Regions, Genetic/genetics , Rhodopsin/biosynthesis , Rhodopsin/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism
11.
J Neurosci ; 35(20): 8004-20, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995483

ABSTRACT

Amacrine interneurons, which are highly diversified in morphological, neurochemical, and physiological features, play crucial roles in visual information processing in the retina. However, the specification mechanisms and functions in vision for each amacrine subtype are not well understood. We found that the Prdm13 transcriptional regulator is specifically expressed in developing and mature amacrine cells in the mouse retina. Most Prdm13-positive amacrine cells are Calbindin- and Calretinin-positive GABAergic or glycinergic neurons. Absence of Prdm13 significantly reduces GABAergic and glycinergic amacrines, resulting in a specific defect of the S2/S3 border neurite bundle in the inner plexiform layer. Forced expression of Prdm13 distinctively induces GABAergic and glycinergic amacrine cells but not cholinergic amacrine cells, whereas Ptf1a, an upstream transcriptional regulator of Prdm13, induces all of these subtypes. Moreover, Prdm13-deficient mice showed abnormally elevated spatial, temporal, and contrast sensitivities in vision. Together, these results show that Prdm13 regulates development of a subset of amacrine cells, which newly defines an amacrine subtype to negatively modulate visual sensitivities. Our current study provides new insights into mechanisms of the diversification of amacrine cells and their function in vision.


Subject(s)
Amacrine Cells/metabolism , Cholinergic Neurons/metabolism , Contrast Sensitivity , Histone-Lysine N-Methyltransferase/metabolism , Interneurons/metabolism , Transcription Factors/metabolism , Amacrine Cells/cytology , Amacrine Cells/physiology , Animals , Cholinergic Neurons/cytology , Cholinergic Neurons/physiology , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Histone-Lysine N-Methyltransferase/genetics , Interneurons/cytology , Interneurons/physiology , Mice , Neurogenesis , Transcription Factors/genetics
12.
Cell Rep ; 10(5): 796-808, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25660028

ABSTRACT

In vertebrate retinal development, the axonal terminals of retinal neurons make synaptic contacts within narrow fixed regions, and these locations are maintained thereafter. However, the mechanisms and biological logic of the organization of these fixed synapse locations are poorly understood. We show here that a membrane scaffold protein, 4.1G, is highly expressed in retinal photoreceptors and is essential for the arrangement of their correct synapse location. The 4.1G-deficient retina exhibits mislocalization of photoreceptor terminals, although their synaptic connections are normally formed. The 4.1G protein binds to the AP3B2 protein, which is involved in neuronal membrane trafficking, and promotes neurite extension in an AP3B2-dependent manner. 4.1G mutant mice showed visual acuity impairments in an optokinetic response, suggesting that correct synapse location is required for normal visual function. Taken together, the data in this study provide insight into the mechanism and importance of proper synapse location in neural circuit formation.

13.
PLoS One ; 8(1): e54146, 2013.
Article in English | MEDLINE | ID: mdl-23335994

ABSTRACT

BACKGROUND: Adeno-associated virus (AAV) is well established as a vehicle for in vivo gene transfer into the mammalian retina. This virus is promising not only for gene therapy of retinal diseases, but also for in vivo functional analysis of retinal genes. Previous reports have shown that AAV can infect various cell types in the developing mouse retina. However, AAV tropism in the developing retina has not yet been examined in detail. METHODOLOGY/PRINCIPAL FINDINGS: We subretinally delivered seven AAV serotypes (AAV2/1, 2/2, 2/5, 2/8, 2/9, 2/10, and 2/11) of AAV-CAG-mCherry into P0 mouse retinas, and quantitatively evaluated the tropisms of each serotype by its infecting degree in retinal cells. After subretinal injection of AAV into postnatal day 0 (P0) mouse retinas, various retinal cell types were efficiently transduced with different AAVs. Photoreceptor cells were efficiently transduced with AAV2/5. Retinal cells, except for bipolar and Müller glial cells, were efficiently transduced with AAV2/9. Horizontal and/or ganglion cells were efficiently transduced with AAV2/1, AAV2/2, AAV2/8, AAV2/9 and AAV2/10. To confirm the usefulness of AAV-mediated gene transfer into the P0 mouse retina, we performed AAV-mediated rescue of the Cone-rod homeobox gene knockout (Crx KO) mouse, which exhibits an outer segment formation defect, flat electroretinogram (ERG) responses, and photoreceptor degeneration. We injected an AAV expressing Crx under the control of the Crx 2kb promoter into the neonatal Crx KO retina. We showed that AAV mediated-Crx expression significantly decreased the abnormalities of the Crx KO retina. CONCLUSION/SIGNIFICANCE: In the current study, we report suitable AAV tropisms for delivery into the developing mouse retina. Using AAV2/5 in photoreceptor cells, we demonstrated the possibility of gene replacement for the developmental disorder and subsequent degeneration of retinal photoreceptors caused by the absence of Crx.


Subject(s)
Dependovirus/physiology , Gene Transfer Techniques , Genetic Vectors , Photoreceptor Cells/metabolism , Retina/metabolism , Viral Tropism , Animals , Dependovirus/classification , Electroretinography , Gene Expression , Gene Knockout Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Photoreceptor Cells/virology , Retina/virology , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Trans-Activators/genetics , Trans-Activators/metabolism , Transduction, Genetic
14.
J Neurosci ; 32(49): 17658-70, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23223288

ABSTRACT

In vertebrate retinal development, various transcription factors are known to execute essential activities in gene regulation. Although epigenetic modification is considered to play a pivotal role in retinal development, the exact in vivo role of epigenetic regulation is still poorly understood. We observed that G9a histone methyltransferase, which methylates histone H3 at lysine 9 (H3K9), is substantially expressed in the mouse retina throughout development. To address in vivo G9a function in the mouse retina, we ablated G9a in retinal progenitor cells by conditional gene knock-out (G9a Dkk3 CKO). The G9a Dkk3 CKO retina exhibited severe morphological defects, including photoreceptor rosette formation, a partial loss of the outer nuclear layer, elevated cell death, and persistent cell proliferation. Progenitor cell-related genes, including several cyclins, Hes1, Chx10, and Lhx2, are methylated on histone H3K9 in the wild-type retina, but they were defective in H3K9 methylation and improperly upregulated at late developmental stages in the G9a Dkk3 CKO retina. Notably, conditional depletion of G9a in postmitotic photoreceptor precursors (G9a Crx CKO) led to the development of an almost normal retina, indicating that G9a activity mainly in retinal progenitor cells, but not in photoreceptor precursors, is essential for normal terminal differentiation of and survival of the retina. Our results suggest that proper epigenetic marks in progenitor cells are important for subsequent appropriate terminal differentiation and survival of retinal cells by repressing progenitor cell-related genes in differentiating retinal cells.


Subject(s)
Cell Death/physiology , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Histone-Lysine N-Methyltransferase/physiology , Retina/growth & development , Stem Cells/enzymology , Animals , Cell Death/genetics , Cell Differentiation/genetics , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/genetics , Histone-Lysine N-Methyltransferase/genetics , Methylation , Mice , Mice, Knockout , Mice, Transgenic , Retina/anatomy & histology , Retina/enzymology , Stem Cells/physiology
15.
J Neurosci ; 31(46): 16792-807, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090505

ABSTRACT

The molecular mechanisms underlying cell fate determination from common progenitors in the vertebrate CNS remain elusive. We previously reported that the OTX2 homeoprotein regulates retinal photoreceptor cell fate determination. While Otx2 transactivation is a pivotal process for photoreceptor cell fate determination, its transactivation mechanism in the retina is unknown. Here, we identified an evolutionarily conserved Otx2 enhancer of ∼500 bp, named embryonic enhancer locus for photoreceptor Otx2 transcription (EELPOT), which can recapitulate initial Otx2 expression in the embryonic mouse retina. We found that the RAX homeoprotein interacts with EELPOT to transactivate Otx2, mainly in the final cell cycle of retinal progenitors. Conditional inactivation of Rax results in downregulation of Otx2 expression in vivo. We also showed that NOTCH-HES signaling negatively regulates EELPOT to suppress Otx2 expression. These results suggest that the integrated activity of cell-intrinsic and -extrinsic factors on EELPOT underlies the molecular basis of photoreceptor cell fate determination in the embryonic retina.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Otx Transcription Factors/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bromodeoxyuridine/metabolism , Cell Cycle/genetics , Cell Differentiation , Chromatin Immunoprecipitation , Embryo, Mammalian , Eye Proteins/genetics , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Organ Culture Techniques , Pregnancy , RNA, Messenger/metabolism , Retina/cytology , Stem Cells/physiology , Time Factors , Trans-Activators/genetics , Transcription Factors/genetics , Transfection/methods
16.
Nat Neurosci ; 14(9): 1125-34, 2011 Aug 21.
Article in English | MEDLINE | ID: mdl-21857657

ABSTRACT

MicroRNA-124a (miR-124a) is the most abundant microRNA expressed in the vertebrate CNS. Despite past investigations into the role of miR-124a, inconsistent results have left the in vivo function of miR-124a unclear. We examined the in vivo function of miR-124a by targeted disruption of Rncr3 (retinal non-coding RNA 3), the dominant source of miR-124a. Rncr3(-/-) mice exhibited abnormalities in the CNS, including small brain size, axonal mis-sprouting of dentate gyrus granule cells and retinal cone cell death. We found that Lhx2 is an in vivo target mRNA of miR-124a. We also observed that LHX2 downregulation by miR-124a is required for the prevention of apoptosis in the developing retina and proper axonal development of hippocampal neurons. These results suggest that miR-124a is essential for the maturation and survival of dentate gyrus neurons and retinal cones, as it represses Lhx2 translation.


Subject(s)
Axons/physiology , Hippocampus/cytology , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Neurons/cytology , Retinal Cone Photoreceptor Cells/physiology , Transcription Factors/metabolism , Analysis of Variance , Animals , Animals, Newborn , Brain/abnormalities , Cell Differentiation/genetics , Cell Survival/genetics , Electroporation/methods , Electroretinography/methods , Embryo, Mammalian , Evoked Potentials, Visual/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , In Situ Nick-End Labeling/methods , LIM-Homeodomain Proteins , Mice , Mice, Transgenic , MicroRNAs/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Untranslated/genetics , Retina/cytology , Retina/physiology , Tissue Culture Techniques , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
PLoS One ; 6(5): e19911, 2011.
Article in English | MEDLINE | ID: mdl-21611200

ABSTRACT

BACKGROUND: Paraneoplastic retinopathy (PR), including cancer-associated retinopathy (CAR) and melanoma-associated retinopathy (MAR), is a progressive retinal disease caused by antibodies generated against neoplasms not associated with the eye. While several autoantibodies against retinal antigens have been identified, there has been no known autoantibody reacting specifically against bipolar cell antigens in the sera of patients with PR. We previously reported that the transient receptor potential cation channel, subfamily M, member 1 (TRPM1) is specifically expressed in retinal ON bipolar cells and functions as a component of ON bipolar cell transduction channels. In addition, this and other groups have reported that human TRPM1 mutations are associated with the complete form of congenital stationary night blindness. The purpose of the current study is to investigate whether there are autoantibodies against TRPM1 in the sera of PR patients exhibiting ON bipolar cell dysfunction. METHODOLOGY/PRINCIPAL FINDINGS: We performed Western blot analysis to identify an autoantibody against TRPM1 in the serum of a patient with lung CAR. The electroretinograms of this patient showed a severely reduced ON response with normal OFF response, indicating that the defect is in the signal transmission between photoreceptors and ON bipolar cells. We also investigated the sera of 26 patients with MAR for autoantibodies against TRPM1 because MAR patients are known to exhibit retinal ON bipolar cell dysfunction. Two of the patients were found to have autoantibodies against TRPM1 in their sera. CONCLUSION/SIGNIFICANCE: Our study reveals TRPM1 to be one of the autoantigens targeted by autoantibodies in at least some patients with CAR or MAR associated with retinal ON bipolar cell dysfunction.


Subject(s)
Autoantibodies/immunology , Paraneoplastic Syndromes/immunology , Paraneoplastic Syndromes/physiopathology , Retinal Bipolar Cells/pathology , Retinal Diseases/immunology , Retinal Diseases/physiopathology , TRPM Cation Channels/immunology , Aged , Blotting, Western , Electroretinography , Humans , Male , Melanoma/blood , Paraneoplastic Syndromes/blood , Paraneoplastic Syndromes/complications , Retinal Diseases/blood , Retinal Diseases/complications , TRPM Cation Channels/blood
18.
Mol Vis ; 16: 425-37, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20300565

ABSTRACT

PURPOSE: To identify human transient receptor potential cation channel, subfamily M, member 1 (TRPM1) gene mutations in patients with congenital stationary night blindness (CSNB). METHODS: We analyzed four different Japanese patients with complete CSNB in whom previous molecular examination revealed no mutation in either nyctalopin (NYX) or glutamate receptor, metabotropic 6 (GRM6). The ophthalmologic examination included best-corrected visual acuity, refraction, biomicroscopy, ophthalmoscopy, fundus photography, Goldmann kinetic perimetry, color vision tests, and electroretinography (ERG). Exons 2 through 27 and the exon-intron junction regions of human TRPM1 were sequenced. RESULTS: Five different mutations in human TRPM1 were identified. Mutations were present in three unrelated patients with complete CSNB. All three patients were compound heterozygotes. Fundus examination revealed no abnormalities other than myopic changes, and the single bright-flash, mixed rod-cone ERG showed a "negative-type" configuration with a reduced normal a-wave and a significantly reduced b-wave amplitude. Our biochemical and cell biologic analyses suggest that the two identified IVS mutations lead to abnormal TRPM1 protein production, and imply that the two identified missense mutations lead to the mislocalization of the TRPM1 protein in bipolar cells (BCs). CONCLUSIONS: Human TRPM1 mutations are associated with the complete form of CSNB in Japanese patients, suggesting that TRPM1 plays an essential role in mediating the photoresponse in ON BCs in humans as well as in mice.


Subject(s)
Genetic Predisposition to Disease , Mutation/genetics , Night Blindness/congenital , Night Blindness/genetics , TRPM Cation Channels/genetics , Adult , Animals , Base Sequence , Cell Line , Child , DNA Mutational Analysis , Electroretinography , Female , Heterozygote , Humans , Intracellular Space/metabolism , Male , Mice , Mice, Knockout , Molecular Sequence Data , Mutation, Missense/genetics , Pedigree , Protein Transport , TRPM Cation Channels/deficiency , TRPM Cation Channels/metabolism , Young Adult
19.
Proc Natl Acad Sci U S A ; 107(1): 332-7, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19966281

ABSTRACT

An essential step in intricate visual processing is the segregation of visual signals into ON and OFF pathways by retinal bipolar cells (BCs). Glutamate released from photoreceptors modulates the photoresponse of ON BCs via metabotropic glutamate receptor 6 (mGluR6) and G protein (Go) that regulates a cation channel. However, the cation channel has not yet been unequivocally identified. Here, we report a mouse TRPM1 long form (TRPM1-L) as the cation channel. We found that TRPM1-L localization is developmentally restricted to the dendritic tips of ON BCs in colocalization with mGluR6. TRPM1 null mutant mice completely lose the photoresponse of ON BCs but not that of OFF BCs. In the TRPM1-L-expressing cells, TRPM1-L functions as a constitutively active nonselective cation channel and its activity is negatively regulated by Go in the mGluR6 cascade. These results demonstrate that TRPM1-L is a component of the ON BC transduction channel downstream of mGluR6 in ON BCs.


Subject(s)
Light Signal Transduction/physiology , Protein Isoforms/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/physiology , TRPM Cation Channels/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Electrophysiology , Humans , Light , Mice , Mice, Knockout , Molecular Sequence Data , Open Reading Frames , Protein Isoforms/genetics , Receptors, Metabotropic Glutamate/genetics , Retinal Bipolar Cells/cytology , TRPM Cation Channels/genetics
20.
FEBS Lett ; 584(4): 753-8, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20026326

ABSTRACT

Neuronal gene transcription is regulated by both transcriptional activators and repressors. While the roles of transactivators in retinal photoreceptor development have been well characterized, the roles of repressors have been poorly understood. We isolated Panky/Ankrd33, a gene encoding an ankyrin repeat-containing protein. Panky-A was specifically expressed in retinal photoreceptors and the pineal gland, and its expression was directly up-regulated by the CRX transcription factor. Subcellular localization of PANKY-A was observed in the nucleus and cytoplasm. Additionally, transactivation analysis suggested that PANKY-A is a transcriptional cofactor that suppresses CRX-activated photoreceptor genes. Furthermore, we found by an electrophoretic mobility shift assay that PANKY inhibited the DNA-binding activity of CRX.


Subject(s)
Ankyrin Repeat , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cell Line , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Gene Expression Profiling , Gene Expression Regulation , Homeodomain Proteins/genetics , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/metabolism , Phylogeny , Repressor Proteins/classification , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Trans-Activators/genetics , Transcription Factors/classification , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...