Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Development ; 147(11)2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32467238

RESUMO

Gene targeting is an incredibly valuable technique. Sometimes, however, it can also be extremely challenging for various intrinsic reasons (e.g. low target accessibility or nature/extent of gene modification). To bypass these barriers, we designed a transgene-based system in Drosophila that increases the number of independent gene targeting events while at the same time enriching for correctly targeted progeny. Unfortunately, with particularly challenging gene targeting experiments, our original design yielded numerous false positives. Here, we deliver a much-improved technique, named Enhanced Golic+ (E-Golic+). E-Golic+ incorporates genetic modifications to tighten lethality-based selection while simultaneously boosting efficiency. With E-Golic+, we easily achieve previously unattainable gene targeting. Additionally, we built an E-Golic+-based, high-efficiency genetic pipeline for transgene swapping. We demonstrate its utility by transforming GAL4 enhancer-trap lines into tissue-specific Cas9-expressing lines. Given the superior efficiency, specificity and scalability, E-Golic+ promises to expedite development of additional sophisticated genetic/genomic tools in Drosophila.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Drosophila/metabolismo , Marcação de Genes/métodos , Transgenes/genética , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Células Germinativas/citologia , Células Germinativas/metabolismo , Masculino , Regiões Promotoras Genéticas , RNA Guia de Cinetoplastídeos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Nucleic Acids Res ; 48(8): 4344-4356, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32187363

RESUMO

The genome is the blueprint for an organism. Interrogating the genome, especially locating critical cis-regulatory elements, requires deletion analysis. This is conventionally performed using synthetic constructs, making it cumbersome and non-physiological. Thus, we created Cas9-mediated Arrayed Mutagenesis of Individual Offspring (CAMIO) to achieve comprehensive analysis of a targeted region of native DNA. CAMIO utilizes CRISPR that is spatially restricted to generate independent deletions in the intact Drosophila genome. Controlled by recombination, a single guide RNA is stochastically chosen from a set targeting a specific DNA region. Combining two sets increases variability, leading to either indels at 1-2 target sites or inter-target deletions. Cas9 restriction to male germ cells elicits autonomous double-strand-break repair, consequently creating offspring with diverse mutations. Thus, from a single population cross, we can obtain a deletion matrix covering a large expanse of DNA at both coarse and fine resolution. We demonstrate the ease and power of CAMIO by mapping 5'UTR sequences crucial for chinmo's post-transcriptional regulation.


Assuntos
Sistemas CRISPR-Cas , Drosophila/genética , Edição de Genes , Mutagênese , Regiões 5' não Traduzidas , Animais , Animais Geneticamente Modificados , Proteína 9 Associada à CRISPR , Proteínas de Drosophila/genética , Genoma de Inseto , Mutação INDEL , Masculino , Proteínas do Tecido Nervoso/genética , Espermatozoides/metabolismo
3.
Development ; 145(11)2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29764857

RESUMO

Macroglial cells in the central nervous system exhibit regional specialization and carry out region-specific functions. Diverse glial cells arise from specific progenitors in specific spatiotemporal patterns. This raises an interesting possibility that glial precursors with distinct developmental fates exist that govern region-specific gliogenesis. Here, we have mapped the glial progeny produced by the Drosophila type II neuroblasts, which, like vertebrate radial glia cells, yield both neurons and glia via intermediate neural progenitors (INPs). Distinct type II neuroblasts produce different characteristic sets of glia. A single INP can make both astrocyte-like and ensheathing glia, which co-occupy a relatively restrictive subdomain. Blocking apoptosis uncovers further lineage distinctions in the specification, proliferation and survival of glial precursors. Both the switch from neurogenesis to gliogenesis and the subsequent glial expansion depend on Notch signaling. Taken together, lineage origins preconfigure the development of individual glial precursors with involvement of serial Notch actions in promoting gliogenesis.


Assuntos
Encéfalo/embriologia , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Receptores Notch/metabolismo , Animais , Apoptose/fisiologia , Astrócitos/citologia , Encéfalo/citologia , Linhagem da Célula/fisiologia , Proliferação de Células/fisiologia , Neurônios/citologia
4.
Development ; 144(19): 3454-3464, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28851709

RESUMO

The termination of the proliferation of Drosophila neural stem cells, also known as neuroblasts (NBs), requires a 'decommissioning' phase that is controlled in a lineage-specific manner. Most NBs, with the exception of those of the mushroom body (MB), are decommissioned by the ecdysone receptor and mediator complex, causing them to shrink during metamorphosis, followed by nuclear accumulation of Prospero and cell cycle exit. Here, we demonstrate that the levels of Imp and Syp RNA-binding proteins regulate NB decommissioning. Descending Imp and ascending Syp expression have been shown to regulate neuronal temporal fate. We show that Imp levels decline slower in the MB than in other central brain NBs. MB NBs continue to express Imp into pupation, and the presence of Imp prevents decommissioning partly by inhibiting the mediator complex. Late-larval induction of transgenic Imp prevents many non-MB NBs from decommissioning in early pupae. Moreover, the presence of abundant Syp in aged NBs permits Prospero accumulation that, in turn, promotes cell cycle exit. Together, our results reveal that progeny temporal fate and progenitor decommissioning are co-regulated in protracted neuronal lineages.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Animais Geneticamente Modificados , Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Larva/metabolismo , Complexo Mediador/metabolismo , Modelos Biológicos , Corpos Pedunculados/citologia , Corpos Pedunculados/metabolismo , Células-Tronco Neurais/citologia , Ligação Proteica , Pupa/metabolismo , Proteínas de Ligação a RNA/genética
5.
Proc Natl Acad Sci U S A ; 114(38): E8091-E8099, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28874527

RESUMO

In their classic experiments, Olds and Milner showed that rats learn to lever press to receive an electric stimulus in specific brain regions. This led to the identification of mammalian reward centers. Our interest in defining the neuronal substrates of reward perception in the fruit fly Drosophila melanogaster prompted us to develop a simpler experimental approach wherein flies could implement behavior that induces self-stimulation of specific neurons in their brains. The high-throughput assay employs optogenetic activation of neurons when the fly occupies a specific area of a behavioral chamber, and the flies' preferential occupation of this area reflects their choosing to experience optogenetic stimulation. Flies in which neuropeptide F (NPF) neurons are activated display preference for the illuminated side of the chamber. We show that optogenetic activation of NPF neuron is rewarding in olfactory conditioning experiments and that the preference for NPF neuron activation is dependent on NPF signaling. Finally, we identify a small subset of NPF-expressing neurons located in the dorsomedial posterior brain that are sufficient to elicit preference in our assay. This assay provides the means for carrying out unbiased screens to map reward neurons in flies.


Assuntos
Proteínas de Drosophila/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Neuropeptídeos/genética
6.
Development ; 143(3): 411-21, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26700685

RESUMO

A brain consists of numerous distinct neurons arising from a limited number of progenitors, called neuroblasts in Drosophila. Each neuroblast produces a specific neuronal lineage. To unravel the transcriptional networks that underlie the development of distinct neuroblast lineages, we marked and isolated lineage-specific neuroblasts for RNA sequencing. We labeled particular neuroblasts throughout neurogenesis by activating a conditional neuroblast driver in specific lineages using various intersection strategies. The targeted neuroblasts were efficiently recovered using a custom-built device for robotic single-cell picking. Transcriptome analysis of mushroom body, antennal lobe and type II neuroblasts compared with non-selective neuroblasts, neurons and glia revealed a rich repertoire of transcription factors expressed among neuroblasts in diverse patterns. Besides transcription factors that are likely to be pan-neuroblast, many transcription factors exist that are selectively enriched or repressed in certain neuroblasts. The unique combinations of transcription factors present in different neuroblasts may govern the diverse lineage-specific neuron fates.


Assuntos
Linhagem da Célula/genética , Drosophila melanogaster/genética , Marcação de Genes , Neurônios/citologia , Robótica , Transcriptoma/genética , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Regulação da Expressão Gênica no Desenvolvimento , Análise de Sequência de RNA , Análise de Célula Única , Fatores de Transcrição/metabolismo
7.
Development ; 141(2): 253-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24306106

RESUMO

Drosophila type II neuroblasts (NBs), like mammalian neural stem cells, deposit neurons through intermediate neural progenitors (INPs) that can each produce a series of neurons. Both type II NBs and INPs exhibit age-dependent expression of various transcription factors, potentially specifying an array of diverse neurons by combinatorial temporal patterning. Not knowing which mature neurons are made by specific INPs, however, conceals the actual variety of neuron types and limits further molecular studies. Here we mapped neurons derived from specific type II NB lineages and found that sibling INPs produced a morphologically similar but temporally regulated series of distinct neuron types. This suggests a common fate diversification program operating within each INP that is modulated by NB age to generate slightly different sets of diverse neurons based on the INP birth order. Analogous mechanisms might underlie the expansion of neuron diversity via INPs in mammalian brain.


Assuntos
Drosophila/citologia , Drosophila/crescimento & desenvolvimento , Células-Tronco Neurais/citologia , Animais , Animais Geneticamente Modificados , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Linhagem da Célula , Drosophila/genética , Modelos Neurológicos , Células-Tronco Neurais/classificação , Células-Tronco Neurais/metabolismo , Neurogênese
8.
Proc Natl Acad Sci U S A ; 111(29): E2967-76, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002478

RESUMO

The type II clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) system has emerged recently as a powerful method to manipulate the genomes of various organisms. Here, we report a toolbox for high-efficiency genome engineering of Drosophila melanogaster consisting of transgenic Cas9 lines and versatile guide RNA (gRNA) expression plasmids. Systematic evaluation reveals Cas9 lines with ubiquitous or germ-line-restricted patterns of activity. We also demonstrate differential activity of the same gRNA expressed from different U6 snRNA promoters, with the previously untested U6:3 promoter giving the most potent effect. An appropriate combination of Cas9 and gRNA allows targeting of essential and nonessential genes with transmission rates ranging from 25-100%. We also demonstrate that our optimized CRISPR/Cas tools can be used for offset nicking-based mutagenesis. Furthermore, in combination with oligonucleotide or long double-stranded donor templates, our reagents allow precise genome editing by homology-directed repair with rates that make selection markers unnecessary. Last, we demonstrate a novel application of CRISPR/Cas-mediated technology in revealing loss-of-function phenotypes in somatic cells following efficient biallelic targeting by Cas9 expressed in a ubiquitous or tissue-restricted manner. Our CRISPR/Cas tools will facilitate the rapid evaluation of mutant phenotypes of specific genes and the precise modification of the genome with single-nucleotide precision. Our results also pave the way for high-throughput genetic screening with CRISPR/Cas.


Assuntos
Sistemas CRISPR-Cas/genética , Drosophila melanogaster/genética , Engenharia Genética , Genoma de Inseto/genética , Células Germinativas/metabolismo , Alelos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Reparo do DNA , Marcação de Genes , Genes Essenciais , Genes de Insetos/genética , Vetores Genéticos , Dados de Sequência Molecular , Mutação/genética , Fenótipo , RNA/genética , Transgenes/genética
9.
PLoS Biol ; 10(11): e1001425, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23185131

RESUMO

Binary cell fate decisions allow the production of distinct sister neurons from an intermediate precursor. Neurons are further diversified based on the birth order of intermediate precursors. Here we examined the interplay between binary cell fate and birth-order-dependent temporal fate in the Drosophila lateral antennal lobe (lAL) neuronal lineage. Single-cell mapping of the lAL lineage by twin-spot mosaic analysis with repressible cell markers (ts-MARCM) revealed that projection neurons (PNs) and local interneurons (LNs) are made in pairs through binary fate decisions. Forty-five types of PNs innervating distinct brain regions arise in a stereotyped sequence; however, the PNs with similar morphologies are not necessarily born in a contiguous window. The LNs are morphologically less diverse than the PNs, and the sequential morphogenetic changes in the two pairs occur independently. Sanpodo-dependent Notch activity promotes and patterns the LN fates. By contrast, Notch diversifies PN temporal fates in a Sanpodo-dispensable manner. These pleiotropic Notch actions underlie the differential temporal fate specification of twin neurons produced by common precursors within a lineage, possibly by modulating postmitotic neurons' responses to Notch-independent transcriptional cascades.


Assuntos
Linhagem da Célula , Proteínas de Drosophila/metabolismo , Drosophila/citologia , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/citologia , Receptores Notch/metabolismo , Animais , Biomarcadores/metabolismo , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Divisão Celular , Drosophila/embriologia , Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Genes de Insetos , Imuno-Histoquímica , Larva/citologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Mecanotransdução Celular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neurônios/metabolismo , Receptores Notch/genética , Análise de Célula Única/métodos
10.
Proc Natl Acad Sci U S A ; 108(33): 13794-9, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21808003

RESUMO

Pavlovian olfactory learning in Drosophila produces two genetically distinct forms of intermediate-term memories: anesthesia-sensitive memory, which requires the amnesiac gene, and anesthesia-resistant memory (ARM), which requires the radish gene. Here, we report that ARM is specifically enhanced or inhibited in flies with elevated or reduced serotonin (5HT) levels, respectively. The requirement for 5HT was additive with the memory defect of the amnesiac mutation but was occluded by the radish mutation. This result suggests that 5HT and Radish protein act on the same pathway for ARM formation. Three supporting lines of evidence indicate that ARM formation requires 5HT released from only two dorsal paired medial (DPM) neurons onto the mushroom bodies (MBs), the olfactory learning and memory center in Drosophila: (i) DPM neurons were 5HT-antibody immunopositive; (ii) temporal inhibition of 5HT synthesis or release from DPM neurons, but not from other serotonergic neurons, impaired ARM formation; (iii) knocking down the expression of d5HT1A serotonin receptors in α/ß MB neurons, which are innervated by DPM neurons, inhibited ARM formation. Thus, in addition to the Amnesiac peptide required for anesthesia-sensitive memory formation, the two DPM neurons also release 5HT acting on MB neurons for ARM formation.


Assuntos
Anestesia/efeitos adversos , Resistência a Medicamentos/genética , Memória/efeitos dos fármacos , Corpos Pedunculados/fisiologia , Serotonina/fisiologia , Animais , Drosophila , Resistência a Medicamentos/fisiologia , Aprendizagem , Memória/fisiologia , Mutação
11.
Development ; 137(1): 43-51, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20023159

RESUMO

Numb can antagonize Notch signaling to diversify the fates of sister cells. We report here that paired sister cells acquire different fates in all three Drosophila neuronal lineages that make diverse types of antennal lobe projection neurons (PNs). Only one in each pair of postmitotic neurons survives into the adult stage in both anterodorsal (ad) and ventral (v) PN lineages. Notably, Notch signaling specifies the PN fate in the vPN lineage but promotes programmed cell death in the missing siblings in the adPN lineage. In addition, Notch/Numb-mediated binary sibling fates underlie the production of PNs and local interneurons from common precursors in the lAL lineage. Furthermore, Numb is needed in the lateral but not adPN or vPN lineages to prevent the appearance of ectopic neuroblasts and to ensure proper self-renewal of neural progenitors. These lineage-specific outputs of Notch/Numb signaling show that a universal mechanism of binary fate decision can be utilized to govern diverse neural sibling differentiations.


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Proteínas de Drosophila/fisiologia , Hormônios Juvenis/fisiologia , Receptores Notch/fisiologia , Transdução de Sinais , Animais , Apoptose/genética , Apoptose/fisiologia , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Hormônios Juvenis/genética , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Fatores do Domínio POU/fisiologia , Receptores Notch/genética
12.
PLoS Biol ; 8(8)2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20808769

RESUMO

Drosophila brains contain numerous neurons that form complex circuits. These neurons are derived in stereotyped patterns from a fixed number of progenitors, called neuroblasts, and identifying individual neurons made by a neuroblast facilitates the reconstruction of neural circuits. An improved MARCM (mosaic analysis with a repressible cell marker) technique, called twin-spot MARCM, allows one to label the sister clones derived from a common progenitor simultaneously in different colors. It enables identification of every single neuron in an extended neuronal lineage based on the order of neuron birth. Here we report the first example, to our knowledge, of complete lineage analysis among neurons derived from a common neuroblast that relay olfactory information from the antennal lobe (AL) to higher brain centers. By identifying the sequentially derived neurons, we found that the neuroblast serially makes 40 types of AL projection neurons (PNs). During embryogenesis, one PN with multi-glomerular innervation and 18 uniglomerular PNs targeting 17 glomeruli of the adult AL are born. Many more PNs of 22 additional types, including four types of polyglomerular PNs, derive after the neuroblast resumes dividing in early larvae. Although different offspring are generated in a rather arbitrary sequence, the birth order strictly dictates the fate of each post-mitotic neuron, including the fate of programmed cell death. Notably, the embryonic progenitor has an altered temporal identity following each self-renewing asymmetric cell division. After larval hatching, the same progenitor produces multiple neurons for each cell type, but the number of neurons for each type is tightly regulated. These observations substantiate the origin-dependent specification of neuron types. Sequencing neuronal lineages will not only unravel how a complex brain develops but also permit systematic identification of neuron types for detailed structure and function analysis of the brain.


Assuntos
Encéfalo/embriologia , Linhagem da Célula , Drosophila/citologia , Técnicas Genéticas , Mosaicismo , Neurogênese , Neurônios/citologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Células Clonais , Drosophila/embriologia , Drosophila/metabolismo , Neurônios/metabolismo , Condutos Olfatórios
13.
Dev Dyn ; 241(1): 57-68, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21932323

RESUMO

Generating diverse neurons in the central nervous system involves three major steps. First, heterogeneous neural progenitors are specified by positional cues at early embryonic stages. Second, neural progenitors sequentially produce neurons or intermediate precursors that acquire different temporal identities based on their birth-order. Third, sister neurons produced during asymmetrical terminal mitoses are given distinct fates. Determining the molecular mechanisms underlying each of these three steps of cellular diversification will unravel brain development and evolution. Drosophila has a relatively simple and tractable CNS, and previous studies on Drosophila CNS development have greatly advanced our understanding of neuron fate specification. Here we review those studies and discuss how the lessons we have learned from fly teach us the process of neuronal diversification in general.


Assuntos
Sistema Nervoso Central/anatomia & histologia , Sistema Nervoso Central/embriologia , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/embriologia , Animais , Linhagem da Célula , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Morfogênese/fisiologia , Neurogênese , Neurônios/citologia , Neurônios/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
14.
Dev Dyn ; 241(1): 169-89, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22174086

RESUMO

BACKGROUND: Phylogenetic footprinting has revealed that cis-regulatory enhancers consist of conserved DNA sequence clusters (CSCs). Currently, there is no systematic approach for enhancer discovery and analysis that takes full-advantage of the sequence information within enhancer CSCs. RESULTS: We have generated a Drosophila genome-wide database of conserved DNA consisting of >100,000 CSCs derived from EvoPrints spanning over 90% of the genome. cis-Decoder database search and alignment algorithms enable the discovery of functionally related enhancers. The program first identifies conserved repeat elements within an input enhancer and then searches the database for CSCs that score highly against the input CSC. Scoring is based on shared repeats as well as uniquely shared matches, and includes measures of the balance of shared elements, a diagnostic that has proven to be useful in predicting cis-regulatory function. To demonstrate the utility of these tools, a temporally-restricted CNS neuroblast enhancer was used to identify other functionally related enhancers and analyze their structural organization. CONCLUSIONS: cis-Decoder reveals that co-regulating enhancers consist of combinations of overlapping shared sequence elements, providing insights into the mode of integration of multiple regulating transcription factors. The database and accompanying algorithms should prove useful in the discovery and analysis of enhancers involved in any developmental process.


Assuntos
Bases de Dados Genéticas , Drosophila melanogaster/genética , Elementos Facilitadores Genéticos , Genoma de Inseto , Algoritmos , Animais , Sequência de Bases , Biologia Computacional/métodos , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Dados de Sequência Molecular , Filogenia , Transgenes
15.
Elife ; 122023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-36695420

RESUMO

Mushroom bodies (MB) of adult Drosophila have a core of thousands of Kenyon neurons; axons of the early-born g class form a medial lobe and those from later-born α'ß' and αß classes form both medial and vertical lobes. The larva, however, hatches with only γ neurons and forms a vertical lobe 'facsimile' using larval-specific axon branches from its γ neurons. MB input (MBINs) and output (MBONs) neurons divide the Kenyon neuron lobes into discrete computational compartments. The larva has 10 such compartments while the adult has 16. We determined the fates of 28 of the 32 MBONs and MBINs that define the 10 larval compartments. Seven compartments are subsequently incorporated into the adult MB; four of their MBINs die, while 12 MBINs/MBONs remodel to function in adult compartments. The remaining three compartments are larval specific. At metamorphosis their MBIN/MBONs trans-differentiate, leaving the MB for other adult brain circuits. The adult vertical lobes are made de novo using MBONs/MBINs recruited from pools of adult-specific neurons. The combination of cell death, compartment shifting, trans-differentiation, and recruitment of new neurons result in no larval MBIN-MBON connections being maintained through metamorphosis. At this simple level, then, we find no anatomical substrate for a memory trace persisting from larva to adult. The adult phenotype of the trans-differentiating neurons represents their evolutionarily ancestral phenotype while their larval phenotype is a derived adaptation for the larval stage. These cells arise primarily within lineages that also produce permanent MBINs and MBONs, suggesting that larval specifying factors may allow information related to birth-order or sibling identity to be interpreted in a modified manner in the larva to allow these neurons to acquire larval phenotypic modifications. The loss of such factors at metamorphosis then allows these neurons to revert to their ancestral functions in the adult.


Assuntos
Drosophila , Neurônios , Animais , Drosophila/fisiologia , Larva/fisiologia , Neurônios/fisiologia , Encéfalo , Axônios , Corpos Pedunculados/fisiologia , Drosophila melanogaster/fisiologia , Metamorfose Biológica
16.
Neuron ; 111(3): 345-361.e10, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36417906

RESUMO

During development, regulatory factors appear in a precise order to determine cell fates over time. Consequently, to investigate complex tissue development, it is necessary to visualize and manipulate cell lineages with temporal control. Current strategies for tracing vertebrate cell lineages lack genetic access to sequentially produced cells. Here, we present TEMPO (Temporal Encoding and Manipulation in a Predefined Order), an imaging-readable genetic tool allowing differential labeling and manipulation of consecutive cell generations in vertebrates. TEMPO is based on CRISPR and powered by a cascade of gRNAs that drive orderly activation and inactivation of reporters and/or effectors. Using TEMPO to visualize zebrafish and mouse neurogenesis, we recapitulated birth-order-dependent neuronal fates. Temporally manipulating cell-cycle regulators in mouse cortex progenitors altered the proportion and distribution of neurons and glia, revealing the effects of temporal gene perturbation on serial cell fates. Thus, TEMPO enables sequential manipulation of molecular factors, crucial to study cell-type specification.


Assuntos
Neurônios , Peixe-Zebra , Animais , Camundongos , Linhagem da Célula/fisiologia , Neurônios/fisiologia , Neuroglia , Diferenciação Celular/genética , Neurogênese/genética , Regulação da Expressão Gênica no Desenvolvimento
17.
Nat Methods ; 6(8): 566-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19644457

RESUMO

Real-time lineage tracing in flies gets a boost with three techniques to specifically label a progenitor's daughter cells.


Assuntos
Linhagem da Célula/genética , Drosophila melanogaster/genética , Técnicas Genéticas , Células-Tronco/citologia , Animais , Drosophila melanogaster/citologia , Proteínas Recombinantes/genética , Transgenes
18.
Adv Exp Med Biol ; 739: 262-75, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22399408

RESUMO

Cell recognition requires interactions through molecules located on cell surface. The insect homolog of Down syndrome cell adhesion molecule (Dscam) manifests huge molecular diversity in its extracellular domain. High-affinity Dscam-Dscam interactions only occur between isoforms that carry identical extracellular domains. Homophilic Dscam signaling can, thus, vary in strength depending on the compositions of Dscams present on the opposing cell surfaces. Dscam abundantly exists in the developing nervous system and governs arborization and proper elaboration of neurites. Notably, individual neurons may stochastically and dynamically express a small subset of Dscam isoforms such that any given neurite can be endowed with a unique repertoire of Dscams. This allows individual neurites to recognize their sister branches. Self-recognition leads to self-repulsion, ensuring divergent migration of sister processes. By contrast, weak homophilic Dscam interactions may promote fasciculation of neurites that express analogous, but not identical, Dscams. Differential Dscam binding may provide graded cell recognition that in turn governs complex neuronal morphogenesis.


Assuntos
Moléculas de Adesão Celular , Comunicação Celular , Animais , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica , Neurônios/citologia , Neurônios/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
19.
Methods Mol Biol ; 2540: 201-217, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35980579

RESUMO

Cell lineage defines the mitotic connection between cells that make up an organism. Mapping these connections in relation to cell identity offers an extraordinary insight into the mechanisms underlying normal and pathological development. The analysis of molecular determinants involved in the acquisition of cell identity requires gaining experimental access to precise parts of cell lineages. Recently, we have developed CaSSA and CLADES, a new technology based on CRISPR that allows targeting and labeling specific lineage branches. Here we discuss how to better exploit this technology for lineage studies in Drosophila, with an emphasis on neuronal specification.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Drosophila , Animais , Linhagem da Célula/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Drosophila/genética , Neurônios/fisiologia
20.
Curr Biol ; 32(10): 2341-2348.e3, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35508173

RESUMO

While we think of neurons as having a fixed identity, many show spectacular plasticity.1-10 Metamorphosis drives massive changes in the fly brain;11,12 neurons that persist into adulthood often change in response to the steroid hormone ecdysone.13,14 Besides driving remodeling,11-14 ecdysone signaling can also alter the differentiation status of neurons.7,15 The three sequentially born subtypes of mushroom body (MB) Kenyon cells (γ, followed by α'/ß', and finally α/ß)16 serve as a model of temporal fating.17-21 γ neurons are also used as a model of remodeling during metamorphosis. As γ neurons are the only functional Kenyon cells in the larval brain, they serve the function of all three adult subtypes. Correspondingly, larval γ neurons have a similar morphology to α'/ß' and α/ß neurons-their axons project dorsally and medially. During metamorphosis, γ neurons remodel to form a single medial projection. Both temporal fate changes and defects in remodeling therefore alter γ-neuron morphology in similar ways. Mamo, a broad-complex, tramtrack, and bric-à-brac/poxvirus and zinc finger (BTB/POZ) transcription factor critical for temporal specification of α'/ß' neurons,18,19 was recently described as essential for γ remodeling.22 In a previous study, we noticed a change in the number of adult Kenyon cells expressing γ-specific markers when mamo was manipulated.18 These data implied a role for Mamo in γ-neuron fate specification, yet mamo is not expressed in γ neurons until pupariation,18,22 well past γ specification. This indicates that mamo has a later role in ensuring that γ neurons express the correct Kenyon cell subtype-specific genes in the adult brain.


Assuntos
Ecdisona , Corpos Pedunculados , Animais , Axônios , Diferenciação Celular , Larva , Corpos Pedunculados/fisiologia , Neurônios/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa