Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Development ; 150(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37721334

ABSTRACT

During neural development, cellular adhesion is crucial for interactions among and between neurons and surrounding tissues. This function is mediated by conserved cell adhesion molecules, which are tightly regulated to allow for coordinated neuronal outgrowth. Here, we show that the proprotein convertase KPC-1 (homolog of mammalian furin) regulates the Menorin adhesion complex during development of PVD dendritic arbors in Caenorhabditis elegans. We found a finely regulated antagonistic balance between PVD-expressed KPC-1 and the epidermally expressed putative cell adhesion molecule MNR-1 (Menorin). Genetically, partial loss of mnr-1 suppressed partial loss of kpc-1, and both loss of kpc-1 and transgenic overexpression of mnr-1 resulted in indistinguishable phenotypes in PVD dendrites. This balance regulated cell-surface localization of the DMA-1 leucine-rich transmembrane receptor in PVD neurons. Lastly, kpc-1 mutants showed increased amounts of MNR-1 and decreased amounts of muscle-derived LECT-2 (Chondromodulin II), which is also part of the Menorin adhesion complex. These observations suggest that KPC-1 in PVD neurons directly or indirectly controls the abundance of proteins of the Menorin adhesion complex from adjacent tissues, thereby providing negative feedback from the dendrite to the instructive cues of surrounding tissues.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Ligands , Cell Membrane , Animals, Genetically Modified , Biological Transport , Caenorhabditis elegans/genetics , Dendrites , Mammals , Membrane Proteins , Caenorhabditis elegans Proteins/genetics
2.
Elife ; 102021 03 17.
Article in English | MEDLINE | ID: mdl-33729157

ABSTRACT

Mutations in the lysine demethylase 5 (KDM5) family of transcriptional regulators are associated with intellectual disability, yet little is known regarding their spatiotemporal requirements or neurodevelopmental contributions. Utilizing the mushroom body (MB), a major learning and memory center within the Drosophila brain, we demonstrate that KDM5 is required within ganglion mother cells and immature neurons for proper axogenesis. Moreover, the mechanism by which KDM5 functions in this context is independent of its canonical histone demethylase activity. Using in vivo transcriptional and binding analyses, we identify a network of genes directly regulated by KDM5 that are critical modulators of neurodevelopment. We find that KDM5 directly regulates the expression of prospero, a transcription factor that we demonstrate is essential for MB morphogenesis. Prospero functions downstream of KDM5 and binds to approximately half of KDM5-regulated genes. Together, our data provide evidence for a KDM5-Prospero transcriptional axis that is essential for proper MB development.


Subject(s)
Drosophila Proteins/metabolism , Histone Demethylases/metabolism , Mushroom Bodies/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Central Nervous System/growth & development , Central Nervous System/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Histone Demethylases/genetics , Larva/growth & development , Larva/metabolism , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics
3.
Cell Rep ; 34(7): 108753, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596422

ABSTRACT

Mutations in the genes encoding the lysine demethylase 5 (KDM5) family of histone demethylases are observed in individuals with intellectual disability (ID). Despite clear evidence linking KDM5 function to neurodevelopmental pathways, how this family of proteins impacts transcriptional programs to mediate synaptic structure and activity remains unclear. Using the Drosophila larval neuromuscular junction (NMJ), we show that KDM5 is required presynaptically for neuroanatomical development and synaptic function. The Jumonji C (JmjC) domain-encoded histone demethylase activity of KDM5, which is expected to be diminished by many ID-associated alleles, is required for appropriate synaptic morphology and neurotransmission. The activity of the C5HC2 zinc finger is also required, as an ID-associated mutation in this motif reduces NMJ bouton number, increases bouton size, and alters microtubule dynamics. KDM5 therefore uses demethylase-dependent and independent mechanisms to regulate NMJ structure and activity, highlighting the complex nature by which this chromatin modifier carries out its neuronal gene-regulatory programs.


Subject(s)
Drosophila Proteins/metabolism , Histone Demethylases/metabolism , Neuromuscular Junction/metabolism , Animals , Drosophila , Female , Male
4.
PLoS One ; 15(7): e0223633, 2020.
Article in English | MEDLINE | ID: mdl-32701951

ABSTRACT

BACKGROUND: Small conductance, calcium-activated (SK3) potassium channels control the intrinsic excitability of dopaminergic neurons (DN) in the midbrain and modulate their susceptibility to toxic insults during development. METHODS: We evaluated the age-dependency of the neuroprotective effect of an SK3 agonist, 1-Ethyl-1,3-dihydro-2H-benzimidazol-2-one (1-EBIO), on Amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) excitotoxicity to DN in ventral mesencephalon (VM) organotypic cultures. RESULTS: Most tyrosine hydroxylase (TH)+ neurons were also SK3+; SK3+/TH- cells (DN+) were common at each developmental stage but more prominently at day in vitro (DIV) 8. Young DN+ neurons were small bipolar and fusiform, whereas mature ones were large and multipolar. Exposure of organotypic cultures to AMPA (100 µm, 16 h) had no effect on the survival of DN+ at DIV 8, but caused significant toxicity at DIV 15 (n = 15, p = 0.005) and DIV 22 (n = 15, p<0.001). These results indicate that susceptibility of DN to AMPA excitotoxicity is developmental stage-dependent in embryonic VM organotypic cultures. Immature DN+ (small, bipolar) were increased after AMPA (100 µm, 16 h) at DIV 8, at the expense of the number of differentiated (large, multipolar) DN+ (p = 0.039). This effect was larger at DIV 15 (p<<<0.0001) and at DIV 22 (p<<<0.0001). At DIV 8, 30 µM 1-EBIO resulted in a large increase in DN+. At DIV 15, AMPA toxicity was prevented by exposure to 30 µM, but not 100 µM 1-EBIO. At DIV 22, excitotoxicity was unaffected by 30 µM 1-EBIO, and partially reduced by 100 µM 1-EBIO. CONCLUSION: The effects of the SK3 channel agonist 1-EBIO on the survival of SK3-expressing dopaminergic neurons were concentration-dependent and influenced by neuronal developmental stage.


Subject(s)
Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/agonists , Animals , Benzimidazoles/pharmacology , Cell Survival/drug effects , Cells, Cultured , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Embryo, Mammalian/cytology , Embryonic Development/drug effects , Female , Mesencephalon/cytology , Mesencephalon/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity
5.
Development ; 146(24)2019 12 20.
Article in English | MEDLINE | ID: mdl-31862793

ABSTRACT

In Drosophila, the larval prothoracic gland integrates nutritional status with developmental signals to regulate growth and maturation through the secretion of the steroid hormone ecdysone. While the nutritional signals and cellular pathways that regulate prothoracic gland function are relatively well studied, the transcriptional regulators that orchestrate the activity of this tissue remain less characterized. Here, we show that lysine demethylase 5 (KDM5) is essential for prothoracic gland function. Indeed, restoring kdm5 expression only in the prothoracic gland in an otherwise kdm5 null mutant animal is sufficient to rescue both the larval developmental delay and the pupal lethality caused by loss of KDM5. Our studies show that KDM5 functions by promoting the endoreplication of prothoracic gland cells, a process that increases ploidy and is rate limiting for the expression of ecdysone biosynthetic genes. Molecularly, we show that KDM5 activates the expression of the receptor tyrosine kinase torso, which then promotes polyploidization and growth through activation of the MAPK signaling pathway. Taken together, our studies provide key insights into the biological processes regulated by KDM5 and expand our understanding of the transcriptional regulators that coordinate animal development.


Subject(s)
Biological Clocks/genetics , Drosophila Proteins/physiology , Drosophila melanogaster , Embryonic Development/genetics , Endocrine Glands/embryology , Histone Demethylases/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Ecdysone/metabolism , Embryo, Nonmammalian , Endocrine Glands/metabolism , Endoreduplication/genetics , Female , Gene Expression Regulation, Developmental , Larva , MAP Kinase Signaling System/physiology , Male , Organogenesis/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Time Factors
6.
Dev Cell ; 48(2): 229-244.e4, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30661986

ABSTRACT

The mechanisms that pattern and maintain dendritic arbors are key to understanding the principles that govern nervous system assembly. The activity of presynaptic axons has long been known to shape dendrites, but activity-independent functions of axons in this process have remained elusive. Here, we show that in Caenorhabditis elegans, the axons of the ALA neuron control guidance and extension of the 1° dendrites of PVD somatosensory neurons independently of ALA activity. PVD 1° dendrites mimic ALA axon guidance defects in loss-of-function mutants for the extracellular matrix molecule MIG-6/Papilin or the UNC-6/Netrin pathway, suggesting that axon-dendrite adhesion is important for dendrite formation. We found that the SAX-7/L1CAM cell adhesion molecule engages in distinct molecular mechanisms to mediate extensions of PVD 1° dendrites and maintain the ALA-PVD axon-dendritic fascicle, respectively. Thus, axons can serve as critical scaffolds to pattern and maintain dendrites through contact-dependent but activity-independent mechanisms.


Subject(s)
Axons/metabolism , Cell Adhesion Molecules/metabolism , Dendrites/metabolism , Neuronal Plasticity/physiology , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism
7.
Genetics ; 209(3): 773-787, 2018 07.
Article in English | MEDLINE | ID: mdl-29764901

ABSTRACT

Regulated gene expression is necessary for developmental and homeostatic processes. The KDM5 family of transcriptional regulators are histone H3 lysine 4 demethylases that can function through both demethylase-dependent and -independent mechanisms. While loss and overexpression of KDM5 proteins are linked to intellectual disability and cancer, respectively, their normal developmental functions remain less characterized. Drosophila melanogaster provides an ideal system to investigate KDM5 function, as it encodes a single ortholog in contrast to the four paralogs found in mammalian cells. To examine the consequences of complete loss of KDM5, we generated a null allele of Drosophila kdm5, also known as little imaginal discs (lid), and show that it is essential for viability. Animals lacking KDM5 show a dramatically delayed larval development that coincides with decreased proliferation and increased cell death in wing imaginal discs. Interestingly, this developmental delay is independent of the well-characterized Jumonji C (JmjC) domain-encoded histone demethylase activity of KDM5, suggesting key functions for less characterized domains. Consistent with the phenotypes observed, transcriptome analyses of kdm5 null mutant wing imaginal discs revealed the dysregulation of genes involved in several cellular processes, including cell cycle progression and DNA repair. Together, our analyses reveal KDM5 as a key regulator of larval growth and offer an invaluable tool for defining the biological activities of KDM5 family proteins.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Regulatory Networks , Histone Demethylases/genetics , Loss of Function Mutation , Wings, Animal/growth & development , Animals , Cell Cycle , Cell Death , Cell Proliferation , DNA Repair , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Histone Demethylases/chemistry , Histone Demethylases/metabolism , Larva/growth & development , Larva/metabolism , Phenotype , Protein Domains , Sequence Analysis, RNA
8.
Cell Rep ; 22(9): 2359-2369, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29490272

ABSTRACT

Mutations in KDM5 family histone demethylases cause intellectual disability in humans. However, the molecular mechanisms linking KDM5-regulated transcription and cognition remain unknown. Here, we establish Drosophila as a model to understand this connection by generating a fly strain harboring an allele analogous to a disease-causing missense mutation in human KDM5C (kdm5A512P). Transcriptome analysis of kdm5A512P flies revealed a striking downregulation of genes required for ribosomal assembly and function and a concomitant reduction in translation. kdm5A512P flies also showed impaired learning and/or memory. Significantly, the behavioral and transcriptional changes in kdm5A512P flies were similar to those specifically lacking demethylase activity. These data suggest that the primary defect of the KDM5A512P mutation is a loss of histone demethylase activity and reveal an unexpected role for this enzymatic function in gene activation. Because translation is critical for neuronal function, we propose that this defect contributes to the cognitive defects of kdm5A512P flies.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Histone Demethylases/genetics , Intellectual Disability/genetics , Mutation/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Cell Nucleus/metabolism , Cognition , Disease Models, Animal , Down-Regulation/genetics , Drosophila Proteins/chemistry , Gene Expression Profiling , Histone Demethylases/chemistry , Histones/metabolism , Lysine/metabolism , Methylation , Phenotype , Promoter Regions, Genetic/genetics , Protein Biosynthesis , Ribosomes/metabolism , Transcription, Genetic , Transcriptional Activation/genetics
9.
Neuropharmacology ; 60(7-8): 1176-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21044638

ABSTRACT

In primary cultures of mesencephalon small-conductance calcium-activated potassium channels (SK) are expressed in dopaminergic neurons. We characterized SK-mediated currents (I(SK)) in this system and evaluated their role on homeostasis against excitotoxicity. I(SK) amplitude was reduced by the glutamatergic agonist AMPA through a reduction in SK channel number in the membrane. Blockade of I(SK) for 12 h with apamin or NS8593 reduced the number of dopaminergic neurons in a concentration-dependent manner. The effect of apamin was not additive to AMPA toxicity. On the other hand, two I(SK) agonists, 1-EBIO and CyPPA, caused a significant reduction of spontaneous loss of dopaminergic neurons. 1-EBIO reversed the effects of both AMPA and apamin as well. Thus, I(SK) influences survival and differentiation of dopaminergic neurons in vitro, and is part of protective homeostatic responses, participating in a rapidly acting negative feedback loop coupling calcium levels, neuron excitability and cellular defenses. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Mesencephalon/drug effects , Neurons/drug effects , Small-Conductance Calcium-Activated Potassium Channels/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/toxicity , Action Potentials/drug effects , Action Potentials/physiology , Animals , Apamin/toxicity , Benzimidazoles/pharmacology , Calcium Channel Agonists/pharmacology , Cell Culture Techniques , Dopamine/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Mesencephalon/metabolism , Neurons/metabolism , Neurotoxins/toxicity , Patch-Clamp Techniques , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, Dopamine/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...