Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell Stem Cell ; 31(3): 283-284, 2024 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-38458174

RESUMO

Dissecting the role of the thalamus in neuropsychiatric disorders requires new models to analyze complex genetic interactions. In this issue of Cell Stem Cell, Shin et al. use patient-derived thalamocortical organoids to investigate 22q11.2 microdeletion impact on thalamic development, revealing significant transcriptional dysregulation linked to psychiatric disorders.


Assuntos
Córtex Cerebral , Transtornos Mentais , Humanos , Vias Neurais , Transtornos Mentais/genética , Tálamo , Organoides
2.
Genetics ; 225(4)2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37793339

RESUMO

Sec1/Munc18 (SM) proteins are important regulators of SNARE complex assembly during exocytosis throughout all major animal tissue types. However, expression of a founding member of the SM family, UNC-18, is mostly restricted to the nervous system of the nematode Caenorhabditis elegans, where it is important for synaptic transmission. Moreover, unc-18 null mutants do not display the lethality phenotype associated with (a) loss of all Drosophila and mouse orthologs of unc-18 and (b) with complete elimination of synaptic transmission in C. elegans. We investigated whether a previously uncharacterized unc-18 paralog, which we named uncp-18, may be able to explain the restricted expression and limited phenotypes of unc-18 null mutants. A reporter allele shows ubiquitous expression of uncp-18. Analysis of uncp-18 null mutants, unc-18 and uncp-18 double null mutants, as well as overexpression of uncp-18 in an unc-18 null mutant background, shows that these 2 genes can functionally compensate for one another and are redundantly required for embryonic viability. Our results indicate that the synaptic transmission defects of unc-18 null mutants cannot necessarily be interpreted as constituting a null phenotype for SM protein function at the synapse.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Camundongos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Mutação , Sinapses/metabolismo , Transmissão Sináptica/genética
3.
Front Cell Neurosci ; 17: 1233830, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37744879

RESUMO

CUT homeobox genes represent a captivating gene class fulfilling critical functions in the development and maintenance of multiple cell types across a wide range of organisms. They belong to the larger group of homeobox genes, which encode transcription factors responsible for regulating gene expression patterns during development. CUT homeobox genes exhibit two distinct and conserved DNA binding domains, a homeodomain accompanied by one or more CUT domains. Numerous studies have shown the involvement of CUT homeobox genes in diverse developmental processes such as body axis formation, organogenesis, tissue patterning and neuronal specification. They govern these processes by exerting control over gene expression through their transcriptional regulatory activities, which they accomplish by a combination of classic and unconventional interactions with the DNA. Intriguingly, apart from their roles as transcriptional regulators, they also serve as accessory factors in DNA repair pathways through protein-protein interactions. They are highly conserved across species, highlighting their fundamental importance in developmental biology. Remarkably, evolutionary analysis has revealed that CUT homeobox genes have experienced an extraordinary degree of rearrangements and diversification compared to other classes of homeobox genes, including the emergence of a novel gene family in vertebrates. Investigating the functions and regulatory networks of CUT homeobox genes provides significant understanding into the molecular mechanisms underlying embryonic development and tissue homeostasis. Furthermore, aberrant expression or mutations in CUT homeobox genes have been associated with various human diseases, highlighting their relevance beyond developmental processes. This review will overview the well known roles of CUT homeobox genes in nervous system development, as well as their functions in other tissues across phylogeny.

4.
Development ; 150(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36595352

RESUMO

Are there common mechanisms of neurogenesis used throughout an entire nervous system? We explored to what extent canonical proneural class I/II bHLH complexes are responsible for neurogenesis throughout the entire Caenorhabditis elegans nervous system. Distinct, lineage-specific proneural class II bHLH factors are generally thought to operate via interaction with a common, class I bHLH subunit, encoded by Daughterless in flies, the E proteins in vertebrates and HLH-2 in C. elegans. To eliminate function of all proneuronal class I/II bHLH complexes, we therefore genetically removed maternal and zygotic hlh-2 gene activity. We observed broad effects on neurogenesis, but still detected normal neurogenesis in many distinct neuron-producing lineages of the central and peripheral nervous system. Moreover, we found that hlh-2 selectively affects some aspects of neuron differentiation while leaving others unaffected. Although our studies confirm the function of proneuronal class I/II bHLH complexes in many different lineages throughout a nervous system, we conclude that their function is not universal, but rather restricted by lineage, cell type and components of differentiation programs affected.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sistema Nervoso/metabolismo , Neurogênese/genética , Regulação da Expressão Gênica no Desenvolvimento
5.
PLoS Genet ; 18(9): e1010372, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36178933

RESUMO

Homeobox genes are prominent regulators of neuronal identity, but the extent to which their function has been probed in animal nervous systems remains limited. In the nematode Caenorhabditis elegans, each individual neuron class is defined by the expression of unique combinations of homeobox genes, prompting the question of whether each neuron class indeed requires a homeobox gene for its proper identity specification. We present here progress in addressing this question by extending previous mutant analysis of homeobox gene family members and describing multiple examples of homeobox gene function in different parts of the C. elegans nervous system. To probe homeobox function, we make use of a number of reporter gene tools, including a novel multicolor reporter transgene, NeuroPAL, which permits simultaneous monitoring of the execution of multiple differentiation programs throughout the entire nervous system. Using these tools, we add to the previous characterization of homeobox gene function by identifying neuronal differentiation defects for 14 homeobox genes in 24 distinct neuron classes that are mostly unrelated by location, function and lineage history. 12 of these 24 neuron classes had no homeobox gene function ascribed to them before, while in the other 12 neuron classes, we extend the combinatorial code of transcription factors required for specifying terminal differentiation programs. Furthermore, we demonstrate that in a particular lineage, homeotic identity transformations occur upon loss of a homeobox gene and we show that these transformations are the result of changes in homeobox codes. Combining the present with past analyses, 113 of the 118 neuron classes of C. elegans are now known to require a homeobox gene for proper execution of terminal differentiation programs. Such broad deployment indicates that homeobox function in neuronal identity specification may be an ancestral feature of animal nervous systems.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Emprego , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox/genética , Neurônios/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Elife ; 112022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35324425

RESUMO

Overarching themes in the terminal differentiation of the enteric nervous system, an autonomously acting unit of animal nervous systems, have so far eluded discovery. We describe here the overall regulatory logic of enteric nervous system differentiation of the nematode Caenorhabditis elegans that resides within the foregut (pharynx) of the worm. A C. elegans homolog of the Drosophila Sine oculis homeobox gene, ceh-34, is expressed in all 14 classes of interconnected pharyngeal neurons from their birth throughout their life time, but in no other neuron type of the entire animal. Constitutive and temporally controlled ceh-34 removal shows that ceh-34 is required to initiate and maintain the neuron type-specific terminal differentiation program of all pharyngeal neuron classes, including their circuit assembly. Through additional genetic loss of function analysis, we show that within each pharyngeal neuron class, ceh-34 cooperates with different homeodomain transcription factors to individuate distinct pharyngeal neuron classes. Our analysis underscores the critical role of homeobox genes in neuronal identity specification and links them to the control of neuronal circuit assembly of the enteric nervous system. Together with the pharyngeal nervous system simplicity as well as its specification by a Sine oculis homolog, our findings invite speculations about the early evolution of nervous systems.


Assuntos
Proteínas de Caenorhabditis elegans , Sistema Nervoso Entérico , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio , Fatores de Transcrição , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , Faringe , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Curr Biol ; 32(8): 1715-1727.e8, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35259341

RESUMO

Pan-neuronally expressed genes, such as genes involved in the synaptic vesicle cycle or in neuropeptide maturation, are critical for proper function of all neurons, but the transcriptional control mechanisms that direct such genes to all neurons of a nervous system remain poorly understood. We show here that six members of the CUT family of homeobox genes control pan-neuronal identity specification in Caenorhabditis elegans. Single CUT mutants show barely any effects on pan-neuronal gene expression or global nervous system function, but such effects become apparent and progressively worsen upon removal of additional CUT family members, indicating a critical role of gene dosage. Overexpression of each individual CUT gene rescued the phenotype of compound mutants, corroborating that gene dosage, rather than the activity of specific members of the gene family, is critical for CUT gene family function. Genome-wide binding profiles, as well as mutation of CUT homeodomain binding sites by CRISPR/Cas9 genome engineering show that CUT genes directly control the expression of pan-neuronal features. Moreover, CUT genes act in conjunction with neuron-type-specific transcription factors to control pan-neuronal gene expression. Our study, therefore, provides a previously missing key insight into how neuronal gene expression programs are specified and reveals a highly buffered and robust mechanism that controls the most critical functional features of all neuronal cell types.


Assuntos
Proteínas de Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/fisiologia
10.
Sci Adv ; 7(15)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33827819

RESUMO

Neural cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterized by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and astrocytes. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across these two brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis reveals the existence of common nucleus-specific progenitors for neurons and astrocytes. Consistent with their shared molecular signature, regional specificity is maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.


Assuntos
Astrócitos , Neocórtex , Astrócitos/metabolismo , Epigenômica , Neurônios/fisiologia , Tálamo
11.
Genetics ; 217(1): 1-17, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33683371

RESUMO

We describe here phase-separated subnuclear organelles in the nematode Caenorhabditis elegans, which we term NUN (NUclear Nervous system-specific) bodies. Unlike other previously described subnuclear organelles, NUN bodies are highly cell type specific. In fully mature animals, 4-10 NUN bodies are observed exclusively in the nucleus of neuronal, glial and neuron-like cells, but not in other somatic cell types. Based on co-localization and genetic loss of function studies, NUN bodies are not related to other previously described subnuclear organelles, such as nucleoli, splicing speckles, paraspeckles, Polycomb bodies, promyelocytic leukemia bodies, gems, stress-induced nuclear bodies, or clastosomes. NUN bodies form immediately after cell cycle exit, before other signs of overt neuronal differentiation and are unaffected by the genetic elimination of transcription factors that control many other aspects of neuronal identity. In one unusual neuron class, the canal-associated neurons, NUN bodies remodel during larval development, and this remodeling depends on the Prd-type homeobox gene ceh-10. In conclusion, we have characterized here a novel subnuclear organelle whose cell type specificity poses the intriguing question of what biochemical process in the nucleus makes all nervous system-associated cells different from cells outside the nervous system.


Assuntos
Espaço Intranuclear/ultraestrutura , Neurônios/ultraestrutura , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Ciclo Celular , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Espaço Intranuclear/metabolismo , Neuroglia/ultraestrutura
12.
Wiley Interdiscip Rev Dev Biol ; 9(4): e374, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32012462

RESUMO

One approach to understand the construction of complex systems is to investigate whether there are simple design principles that are commonly used in building such a system. In the context of nervous system development, one may ask whether the generation of its highly diverse sets of constituents, that is, distinct neuronal cell types, relies on genetic mechanisms that share specific common features. Specifically, are there common patterns in the function of regulatory genes across different neuron types and are those regulatory mechanisms not only used in different parts of one nervous system, but are they conserved across animal phylogeny? We address these questions here by focusing on one specific, highly conserved and well-studied regulatory factor, the POU homeodomain transcription factor UNC-86. Work over the last 30 years has revealed a common and paradigmatic theme of unc-86 function throughout most of the neuron types in which Caenorhabditis elegans unc-86 is expressed. Apart from its role in preventing lineage reiterations during development, UNC-86 operates in combination with distinct partner proteins to initiate and maintain terminal differentiation programs, by coregulating a vast array of functionally distinct identity determinants of specific neuron types. Mouse orthologs of unc-86, the Brn3 genes, have been shown to fulfill a similar function in initiating and maintaining neuronal identity in specific parts of the mouse brain and similar functions appear to be carried out by the sole Drosophila ortholog, Acj6. The terminal selector function of UNC-86 in many different neuron types provides a paradigm for neuronal identity regulation across phylogeny. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Invertebrate Organogenesis > Worms Nervous System Development > Vertebrates: Regional Development.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Fatores do Domínio POU/genética , Fator de Transcrição Brn-3C/genética , Animais , Caenorhabditis elegans/classificação , Caenorhabditis elegans/citologia , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/metabolismo , Diferenciação Celular , Linhagem da Célula/genética , Sequência Conservada , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/classificação , Drosophila melanogaster/citologia , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/citologia , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fatores do Domínio POU/metabolismo , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais , Fator de Transcrição Brn-3C/metabolismo
13.
Development ; 146(13)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31227642

RESUMO

The expression of transcription factors that initiate the specification of a unique cellular identity in multicellular organisms is often maintained throughout the life of the respective cell type via an autoregulatory mechanism. It is generally assumed that such autoregulation serves to maintain the differentiated state of a cell. To experimentally test this assumption, we used CRISPR/Cas9-mediated genome engineering to delete a transcriptional autoregulatory, cis-acting motif in the che-1 zinc-finger transcription factor locus, a terminal selector required to specify the identity of the ASE neuron pair during embryonic development of the nematode Caenorhabditis elegans. We show that che-1 autoregulation is indeed required to maintain the differentiated state of the ASE neurons but that it is also required to amplify che-1 expression during embryonic development to reach an apparent minimal threshold to initiate the ASE differentiation program. We conclude that transcriptional autoregulation fulfills two intrinsically linked purposes: one in proper initiation, the other in proper maintenance of terminal differentiation programs.This article has an associated 'The people behind the papers' interview.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Diferenciação Celular/genética , Homeostase/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neurônios/fisiologia , Fatores de Transcrição/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Embrião não Mamífero , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/genética , Dedos de Zinco/fisiologia
14.
Curr Biol ; 28(17): 2813-2823.e2, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30146154

RESUMO

Many distinct regulatory factors have been shown to be required for the proper initiation of neuron-type-specific differentiation programs, but much less is known about the regulatory programs that maintain the differentiated state in the adult [1-3]. One possibility is that regulatory factors that initiate a terminal differentiation program during development are continuously required to maintain the differentiated state. Here, we test this hypothesis by investigating the function of two orthologous POU homeobox genes in nematodes and mice. The C. elegans POU homeobox gene unc-86 is a terminal selector that is required during development to initiate the terminal differentiation program of several distinct neuron classes [4-13]. Through post-developmental removal of unc-86 activity, we show here that unc-86 is also continuously required throughout the life of many neuron classes to maintain neuron-class-specific identity features. Similarly, the mouse unc-86 ortholog Brn3a/POU4F1 has been shown to control the initiation of the terminal differentiation program of distinct neuron types across the mouse brain, such as the medial habenular neurons [14-20]. By conditionally removing Brn3a in the adult mouse central nervous system, we show that, like its invertebrate ortholog unc-86, Brn3a is also required for the maintenance of terminal identity features of medial habenular neurons. In addition, Brn3a is required for the survival of these neurons, indicating that identity maintenance and survival are genetically linked. We conclude that the continuous expression of transcription factors is essential for the active maintenance of the differentiated state of a neuron across phylogeny.


Assuntos
Caenorhabditis elegans/genética , Diferenciação Celular/fisiologia , Neurônios/fisiologia , Fatores do Domínio POU/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Genes Homeobox , Camundongos , Mitose , Neurogênese , Fatores do Domínio POU/genética , Tamoxifeno/farmacologia , Fatores de Transcrição/metabolismo
15.
Genetics ; 207(2): 529-545, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28801529

RESUMO

Repetitive DNA sequences are subject to gene silencing in various animal species. Under specific circumstances repetitive DNA sequences can escape such silencing. For example, exogenously added, extrachromosomal DNA sequences that are stably inherited in multicopy repetitive arrays in the nematode Caenorhabditis elegans are frequently silenced in the germline, whereas such silencing often does not occur in the soma. This indicates that somatic cells might utilize factors that prevent repetitive DNA silencing. Indeed, such "antisilencing" factors have been revealed through genetic screens that identified mutant loci in which repetitive transgenic arrays are aberrantly silenced in the soma. We describe here a novel locus, pals-22 (for protein containing ALS2CR12 signature), required to prevent silencing of repetitive transgenes in neurons and other somatic tissue types. pals-22 deficiency also severely impacts animal vigor and confers phenotypes reminiscent of accelerated aging. We find that pals-22 is a member of a large family of divergent genes (39 members), defined by homology to the ALS2CR12 protein family. While gene family members are highly divergent, they show striking patterns of chromosomal clustering. The family expansion appears C. elegans-specific and has not occurred to the same extent in other nematode species for which genome sequences are available. The transgene-silencing phenotype observed upon loss of PALS-22 protein depends on the biogenesis of small RNAs. We speculate that the pals gene family may be part of a species-specific cellular defense mechanism.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Inativação Gênica , Família Multigênica , Sequências Repetitivas de Ácido Nucleico , Envelhecimento/genética , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/metabolismo , Neurônios/metabolismo , Fenótipo , Transgenes
16.
J Neurosci ; 35(42): 14205-19, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26490861

RESUMO

The striatum is a large brain nucleus with an important role in the control of movement and emotions. Medium spiny neurons (MSNs) are striatal output neurons forming prominent descending axon tracts that target different brain nuclei. However, how MSN axon tracts in the forebrain develop remains poorly understood. Here, we implicate the Wnt binding receptor Frizzled3 in several uncharacterized aspects of MSN pathway formation [i.e., anterior-posterior guidance of MSN axons in the striatum and their subsequent growth into the globus pallidus (GP), an important (intermediate) target]. In Frizzled3 knock-out mice, MSN axons fail to extend along the anterior-posterior axis of the striatum, and many do not reach the GP. Wnt5a acts as an attractant for MSN axons in vitro, is expressed in a posterior high, anterior low gradient in the striatum, and Wnt5a knock-out mice phenocopy striatal anterior-posterior defects observed in Frizzled3 mutants. This suggests that Wnt5a controls anterior-posterior guidance of MSN axons through Frizzled3. Axons that reach the GP in Frizzled3 knock-out mice fail to enter this structure. Surprisingly, entry of MSN axons into the GP non-cell-autonomously requires Frizzled3, and our data suggest that GP entry may be contingent on the correct positioning of "corridor" guidepost cells for thalamocortical axons by Frizzled3. Together, these data dissect MSN pathway development and reveal (non)cell-autonomous roles for Frizzled3 in MSN axon guidance. Further, they are the first to identify a gene that provides anterior-posterior axon guidance in a large brain nucleus and link Frizzled3 to corridor cell development. SIGNIFICANCE STATEMENT: Striatal axon pathways mediate complex physiological functions and are an important therapeutic target, underscoring the need to define how these connections are established. Remarkably, the molecular programs regulating striatal pathway development remain poorly characterized. Here, we determine the embryonic ontogeny of the two main striatal pathways (striatonigral and striatopallidal) and identify novel (non)cell-autonomous roles for the axon guidance receptor Frizzled3 in uncharacterized aspects of striatal pathway formation (i.e., anterior-posterior axon guidance in the striatum and axon entry into the globus pallidus). Further, our results link Frizzled3 to corridor guidepost cell development and suggest that an abnormal distribution of these cells has unexpected, widespread effects on the development of different axon tracts (i.e., striatal and thalamocortical axons).


Assuntos
Axônios/fisiologia , Polaridade Celular/genética , Corpo Estriado/citologia , Receptores Frizzled/metabolismo , Vias Neurais/embriologia , Neurônios/citologia , Animais , Células Cultivadas , Corpo Estriado/embriologia , Embrião de Mamíferos , Feminino , Receptores Frizzled/genética , Globo Pálido/citologia , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Cell Rep ; 12(9): 1367-76, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26299962

RESUMO

ΒACE1 is the major drug target for Alzheimer's disease, but we know surprisingly little about its normal function in the CNS. Here, we show that this protease is critically involved in semaphorin 3A (Sema3A)-mediated axonal guidance processes in thalamic and hippocampal neurons. An active membrane-bound proteolytic CHL1 fragment is generated by BACE1 upon Sema3A binding. This fragment relays the Sema3A signal via ezrin-radixin-moesin (ERM) proteins to the neuronal cytoskeleton. APH1B-γ-secretase-mediated degradation of this fragment stops the Sema3A-induced collapse and sensitizes the growth cone for the next axonal guidance cue. Thus, we reveal a cycle of proteolytic activity underlying growth cone collapse and restoration used by axons to find their correct trajectory in the brain. Our data also suggest that BACE1 and γ-secretase inhibition have physiologically opposite effects in this process, supporting the idea that combination therapy might attenuate some of the side effects associated with these drugs.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Endopeptidases/metabolismo , Cones de Crescimento/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Células Cultivadas , Endopeptidases/genética , Hipocampo/citologia , Hipocampo/metabolismo , Proteínas de Membrana , Camundongos , Proteólise , Semaforina-3A/genética , Semaforina-3A/metabolismo
18.
Curr Biol ; 24(5): 494-508, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24560577

RESUMO

BACKGROUND: Guidance molecules are normally presented to cells in an overlapping fashion; however, little is known about how their signals are integrated to control the formation of neural circuits. In the thalamocortical system, the topographical sorting of distinct axonal subpopulations relies on the emergent cooperation between Slit1 and Netrin-1 guidance cues presented by intermediate cellular targets. However, the mechanism by which both cues interact to drive distinct axonal responses remains unknown. RESULTS: Here, we show that the attractive response to the guidance cue Netrin-1 is controlled by Slit/Robo1 signaling and by FLRT3, a novel coreceptor for Robo1. While thalamic axons lacking FLRT3 are insensitive to Netrin-1, thalamic axons containing FLRT3 can modulate their Netrin-1 responsiveness in a context-dependent manner. In the presence of Slit1, both Robo1 and FLRT3 receptors are required to induce Netrin-1 attraction by the upregulation of surface DCC through the activation of protein kinase A. Finally, the absence of FLRT3 produces defects in axon guidance in vivo. CONCLUSIONS: These results highlight a novel mechanism by which interactions between limited numbers of axon guidance cues can multiply the responses in developing axons, as required for proper axonal tract formation in the mammalian brain.


Assuntos
Axônios/fisiologia , Glicoproteínas de Membrana/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Receptor DCC , Regulação da Expressão Gênica no Desenvolvimento , Técnicas In Vitro , Glicoproteínas de Membrana/genética , Camundongos Mutantes , Camundongos Transgênicos , Mutação , Fatores de Crescimento Neural/genética , Proteínas do Tecido Nervoso/genética , Netrina-1 , Técnicas de Cultura de Órgãos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/genética , Tálamo/citologia , Tálamo/crescimento & desenvolvimento , Tálamo/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Roundabout
19.
Nat Neurosci ; 15(8): 1134-43, 2012 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-22772332

RESUMO

Developing axons must control their growth rate to follow the appropriate pathways and establish specific connections. However, the regulatory mechanisms involved remain elusive. By combining live imaging with transplantation studies in mice, we found that spontaneous calcium activity in the thalamocortical system and the growth rate of thalamocortical axons were developmentally and intrinsically regulated. Indeed, the spontaneous activity of thalamic neurons governed axon growth and extension through the cortex in vivo. This activity-dependent modulation of growth was mediated by transcriptional regulation of Robo1 through an NF-κB binding site. Disruption of either the Robo1 or Slit1 genes accelerated the progression of thalamocortical axons in vivo, and interfering with Robo1 signaling restored normal axon growth in electrically silent neurons. Thus, modifications to spontaneous calcium activity encode a switch in the axon outgrowth program that allows the establishment of specific neuronal connections through the transcriptional regulation of Slit1 and Robo1 signaling.


Assuntos
Axônios/fisiologia , Sinalização do Cálcio/genética , Córtex Cerebral/fisiologia , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Tálamo/fisiologia , Animais , Axônios/patologia , Cálcio/metabolismo , Córtex Cerebral/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Receptores Imunológicos/fisiologia , Tálamo/crescimento & desenvolvimento , Proteínas Roundabout
20.
Curr Biol ; 21(20): 1748-55, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-22000108

RESUMO

How guidance cues are integrated during the formation of complex axonal tracts remains largely unknown. Thalamocortical axons (TCAs), which convey sensory and motor information to the neocortex, have a rostrocaudal topographic organization initially established within the ventral telencephalon [1-3]. Here, we show that this topography is set in a small hub, the corridor, which contains matching rostrocaudal gradients of Slit1 and Netrin 1. Using in vitro and in vivo experiments, we show that Slit1 is a rostral repellent that positions intermediate axons. For rostral axons, although Slit1 is also repulsive and Netrin 1 has no chemotactic activity, the two factors combined generate attraction. These results show that Slit1 has a dual context-dependent role in TCA pathfinding and furthermore reveal that a combination of cues produces an emergent activity that neither of them has alone. Our study thus provides a novel framework to explain how a limited set of guidance cues can generate a vast diversity of axonal responses necessary for proper wiring of the nervous system.


Assuntos
Axônios/fisiologia , Cones de Crescimento/fisiologia , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Tálamo/embriologia , Tálamo/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Células COS , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Chlorocebus aethiops , Efrina-A5/genética , Efrina-A5/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Fatores de Crescimento Neural/genética , Proteínas do Tecido Nervoso/genética , Netrina-1 , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Proteínas Supressoras de Tumor/genética , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , Proteínas Roundabout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA