Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
mBio ; 14(3): e0051023, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37052506

RESUMO

Microbial components have a range of direct effects on the fetal brain. However, little is known about the cellular targets and molecular mechanisms that mediate these effects. Neural progenitor cells (NPCs) control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. We identify ventricular radial glia (vRG), the primary NPC, as the target of bacterial cell wall (BCW) generated during the antibiotic treatment of maternal pneumonia. BCW enhanced proliferative potential of vRGs by shortening the cell cycle and increasing self-renewal. Expanded vRGs propagated to increase neuronal output in all cortical layers. Remarkably, Toll-like receptor 2 (TLR2), which recognizes BCW, localized at the base of primary cilia in vRGs and the BCW-TLR2 interaction suppressed ciliogenesis leading to derepression of Hedgehog (HH) signaling and expansion of vRGs. We also show that TLR6 is an essential partner of TLR2 in this process. Surprisingly, TLR6 alone was required to set the number of cortical neurons under healthy conditions. These findings suggest that an endogenous signal from TLRs suppresses cortical expansion during normal development of the neocortex and that BCW antagonizes that signal through the TLR2/cilia/HH signaling axis changing brain structure and function. IMPORTANCE Fetal brain development in early gestation can be impacted by transplacental infection, altered metabolites from the maternal microbiome, or maternal immune activation. It is less well understood how maternal microbial subcomponents that cross the placenta, such as bacterial cell wall (BCW), directly interact with fetal neural progenitors and neurons and affect development. This scenario plays out in the clinic when BCW debris released during antibiotic therapy of maternal infection traffics to the fetal brain. This study identifies the direct interaction of BCW with TLR2/6 present on the primary cilium, the signaling hub on fetal neural progenitor cells (NPCs). NPCs control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. Within a window of vulnerability before the appearance of fetal immune cells, the BCW-TLR2/6 interaction results in the inhibition of ciliogenesis, derepression of Sonic Hedgehog signaling, excess proliferation of neural progenitors, and abnormal cortical architecture. In the first example of TLR signaling linked to Sonic Hedgehog, BCW/TLR2/6 appears to act during fetal brain morphogenesis to play a role in setting the total cell number in the neocortex.


Assuntos
Proteínas Hedgehog , Neocórtex , Gravidez , Feminino , Humanos , Proteínas Hedgehog/metabolismo , Neocórtex/metabolismo , Receptor 2 Toll-Like/metabolismo , Ligantes , Receptor 6 Toll-Like/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(38): e2206147119, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36095192

RESUMO

The neocortex, the center for higher brain function, first emerged in mammals and has become massively expanded and folded in humans, constituting almost half the volume of the human brain. Primary microcephaly, a developmental disorder in which the brain is smaller than normal at birth, results mainly from there being fewer neurons in the neocortex because of defects in neural progenitor cells (NPCs). Outer radial glia (oRGs), NPCs that are abundant in gyrencephalic species but rare in lissencephalic species, are thought to play key roles in the expansion and folding of the neocortex. However, how oRGs expand, whether they are necessary for neocortical folding, and whether defects in oRGs cause microcephaly remain important questions in the study of brain development, evolution, and disease. Here, we show that oRG expansion in mice, ferrets, and human cerebral organoids requires cyclin-dependent kinase 6 (CDK6), the mutation of which causes primary microcephaly via an unknown mechanism. In a mouse model in which increased Hedgehog signaling expands oRGs and intermediate progenitor cells and induces neocortical folding, CDK6 loss selectively decreased oRGs and abolished neocortical folding. Remarkably, this function of CDK6 in oRG expansion did not require its kinase activity, was not shared by the highly similar CDK4 and CDK2, and was disrupted by the mutation causing microcephaly. Therefore, our results indicate that CDK6 is conserved to promote oRG expansion, that oRGs are necessary for neocortical folding, and that defects in oRG expansion may cause primary microcephaly.


Assuntos
Quinase 6 Dependente de Ciclina , Células Ependimogliais , Microcefalia , Neocórtex , Animais , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Células Ependimogliais/citologia , Células Ependimogliais/enzimologia , Furões , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Microcefalia/genética , Neocórtex/anormalidades , Neocórtex/enzimologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/enzimologia , Organoides/embriologia
3.
Genes Dev ; 36(11-12): 737-751, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35798383

RESUMO

The primary cilium, a signaling organelle projecting from the surface of a cell, controls cellular physiology and behavior. The presence or absence of primary cilia is a distinctive feature of a given tumor type; however, whether and how the primary cilium contributes to tumorigenesis are unknown for most tumors. Medulloblastoma (MB) is a common pediatric brain cancer comprising four groups: SHH, WNT, group 3 (G3), and group 4 (G4). From 111 cases of MB, we show that primary cilia are abundant in SHH and WNT MBs but rare in G3 and G4 MBs. Using WNT and G3 MB mouse models, we show that primary cilia promote WNT MB by facilitating translation of mRNA encoding ß-catenin, a major oncoprotein driving WNT MB, whereas cilium loss promotes G3 MB by disrupting cell cycle control and destabilizing the genome. Our findings reveal tumor type-specific ciliary functions and underlying molecular mechanisms. Moreover, we expand the function of primary cilia to translation control and reveal a molecular mechanism by which cilia regulate cell cycle progression, thereby providing new frameworks for studying cilium function in normal and pathologic conditions.


Assuntos
Neoplasias Encefálicas , Neoplasias Cerebelares , Meduloblastoma , Animais , Neoplasias Encefálicas/patologia , Ciclo Celular/genética , Neoplasias Cerebelares/genética , Cílios/genética , Humanos , Meduloblastoma/genética , Camundongos
4.
Cereb Cortex ; 31(10): 4730-4741, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34002221

RESUMO

The neocortex, the center for higher brain function, emerged in mammals and expanded in the course of evolution. The expansion of outer radial glia (oRGs) and intermediate progenitor cells (IPCs) plays key roles in the expansion and consequential folding of the neocortex. Therefore, understanding the mechanisms of oRG and IPC expansion is important for understanding neocortical development and evolution. By using mice and human cerebral organoids, we previously revealed that hedgehog (HH) signaling expands oRGs and IPCs. Nevertheless, it remained to be determined whether HH signaling expanded oRGs and IPCs in vivo in gyrencephalic species, in which oRGs and IPCs are naturally expanded. Here, we show that HH signaling is necessary and sufficient to expand oRGs and IPCs in ferrets, a gyrencephalic species, through conserved cellular mechanisms. HH signaling increases oRG-producing division modes of ventricular radial glia (vRGs), oRG self-renewal, and IPC proliferation. Notably, HH signaling affects vRG division modes only in an early restricted phase before superficial-layer neuron production peaks. Beyond this restricted phase, HH signaling promotes oRG self-renewal. Thus, HH signaling expands oRGs and IPCs in two distinct but continuous phases during cortical development.


Assuntos
Córtex Cerebral/fisiologia , Células Ependimogliais/fisiologia , Furões/fisiologia , Proteínas Hedgehog/fisiologia , Transdução de Sinais/fisiologia , Animais , Córtex Cerebral/citologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Neocórtex/crescimento & desenvolvimento , Neocórtex/fisiologia , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Técnicas de Cultura de Órgãos , Gravidez
5.
J Neurosci ; 38(44): 9338-9345, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30381425

RESUMO

Sonic Hedgehog (SHH) signaling has been most widely known for its role in specifying region and cell-type identity during embryonic morphogenesis. This mini-review accompanies a 2018 SFN mini-symposium that addresses an emerging body of research focused on understanding the diverse roles for Shh signaling in a wide range of contexts in neurodevelopment and, more recently, in the mature CNS. Such research shows that Shh affects the function of brain circuits, including the production and maintenance of diverse cell types and the establishment of wiring specificity. Here, we review these novel and unexpected functions and the unanswered questions regarding the role of SHH and its signaling pathway members in these cases.


Assuntos
Proteínas Hedgehog/fisiologia , Morfogênese/fisiologia , Células-Tronco Neurais/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Animais , Humanos , Células-Tronco Neurais/química , Sinapses/química
6.
Am J Pathol ; 188(1): 11-22, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29030052

RESUMO

The primary cilium, a sensory appendage that is present in most mammalian cells, plays critical roles in signaling pathways and cell cycle progression. Mutations that affect the structure or function of primary cilia result in ciliopathies, a group of developmental and degenerative diseases that affect almost all organs and tissues. Our understanding of the constituents, development, and function of primary cilia has advanced considerably in recent years, revealing pathogenic mechanisms that potentially underlie ciliopathies. In the brain, the primary cilia are crucial for early patterning, neurogenesis, neuronal maturation and survival, and tumorigenesis, mostly through regulating cell cycle progression, Hedgehog signaling, and WNT signaling. We review these advances in our knowledge of primary cilia, focusing on brain development, and discuss the mechanisms that may underlie brain abnormalities in ciliopathies.


Assuntos
Encefalopatias/patologia , Encéfalo/patologia , Cílios/patologia , Animais , Encéfalo/crescimento & desenvolvimento , Carcinogênese/patologia , Humanos , Mutação , Neurogênese/fisiologia
7.
Dev Cell ; 43(6): 673-688.e5, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29103956

RESUMO

Mechanistic target of rapamycin (MTOR) cooperates with Hedgehog (HH) signaling, but the underlying mechanisms are incompletely understood. Here we provide genetic, biochemical, and pharmacologic evidence that MTOR complex 1 (mTORC1)-dependent translation is a prerequisite for HH signaling. The genetic loss of mTORC1 function inhibited HH signaling-driven growth of the cerebellum and medulloblastoma. Inhibiting translation or mTORC1 blocked HH signaling. Depleting 4EBP1, an mTORC1 target that inhibits translation, alleviated the dependence of HH signaling on mTORC1. Consistent with this, phosphorylated 4EBP1 levels were elevated in HH signaling-driven medulloblastomas in mice and humans. In mice, an mTORC1 inhibitor suppressed medulloblastoma driven by a mutant SMO that is inherently resistant to existing SMO inhibitors, prolonging the survival of the mice. Our study reveals that mTORC1-mediated translation is a key component of HH signaling and an important target for treating medulloblastoma and other cancers driven by HH signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas de Transporte/antagonistas & inibidores , Neoplasias Cerebelares/metabolismo , Proteínas Hedgehog/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Meduloblastoma/metabolismo , Fosfoproteínas/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Proliferação de Células/fisiologia , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Fatores de Iniciação em Eucariotos , Proteínas Hedgehog/genética , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Meduloblastoma/genética , Meduloblastoma/patologia , Camundongos , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Proteína Gli2 com Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/metabolismo
9.
Nat Commun ; 7: 11876, 2016 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-27336173

RESUMO

Maintenance of epithelial cell polarity and epithelial barrier relies on the spatial organization of the actin cytoskeleton and proper positioning/assembly of intercellular junctions. However, how these processes are regulated is poorly understood. Here we reveal a key role for the multifunctional protein Alix in both processes. In a knockout mouse model of Alix, we identified overt structural changes in the epithelium of the choroid plexus and in the ependyma, such as asymmetrical cell shape and size, misplacement and abnormal beating of cilia, blebbing of the microvilli. These defects culminate in excessive cell extrusion, enlargement of the lateral ventricles and hydrocephalus. Mechanistically, we find that by interacting with F-actin, the Par complex and ZO-1, Alix ensures the formation and maintenance of the apically restricted actomyosin-tight junction complex. We propose that in this capacity Alix plays a role in the establishment of apical-basal polarity and in the maintenance of the epithelial barrier.


Assuntos
Actomiosina/metabolismo , Barreira Hematoencefálica , Proteínas de Ligação ao Cálcio/fisiologia , Plexo Corióideo/metabolismo , Junções Íntimas/metabolismo , Actinas/metabolismo , Animais , Polaridade Celular , Plexo Corióideo/ultraestrutura , Epêndima/ultraestrutura , Células Epiteliais/ultraestrutura , Hidrocefalia/etiologia , Camundongos , Camundongos Knockout , Proteína da Zônula de Oclusão-1/metabolismo
10.
Nat Neurosci ; 19(7): 888-96, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27214567

RESUMO

The unique mental abilities of humans are rooted in the immensely expanded and folded neocortex, which reflects the expansion of neural progenitors, especially basal progenitors including basal radial glia (bRGs) and intermediate progenitor cells (IPCs). We found that constitutively active Sonic hedgehog (Shh) signaling expanded bRGs and IPCs and induced folding in the otherwise smooth mouse neocortex, whereas the loss of Shh signaling decreased the number of bRGs and IPCs and the size of the neocortex. SHH signaling was strongly active in the human fetal neocortex but Shh signaling was not strongly active in the mouse embryonic neocortex, and blocking SHH signaling in human cerebral organoids decreased the number of bRGs. Mechanistically, Shh signaling increased the initial generation and self-renewal of bRGs and IPC proliferation in mice and the initial generation of bRGs in human cerebral organoids. Thus, robust SHH signaling in the human fetal neocortex may contribute to bRG and IPC expansion and neocortical growth and folding.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neocórtex/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas Hedgehog/genética , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo
11.
Cancer Cell ; 29(1): 5-16, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-26766587

RESUMO

Four distinct subgroups of cerebellar medulloblastomas (MBs) differ in their histopathology, molecular profiles, and prognosis. c-Myc (Myc) or MycN overexpression in granule neuron progenitors (GNPs) induces Group 3 (G3) or Sonic Hedgehog (SHH) MBs, respectively. Differences in Myc and MycN transcriptional profiles depend, in part, on their interaction with Miz1, which binds strongly to Myc but not MycN, to target sites on chromatin. Myc suppresses ciliogenesis and reprograms the transcriptome of SHH-dependent GNPs through Miz1-dependent gene repression to maintain stemness. Genetic disruption of the Myc/Miz1 interaction inhibited G3 MB development. Target genes of Myc/Miz1 are repressed in human G3 MBs but not in other subgroups. Therefore, the Myc/Miz1 interaction is a defining hallmark of G3 MB development.


Assuntos
Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Meduloblastoma/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/genética , Camundongos , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases
12.
Neurogenesis (Austin) ; 3(1): e1242957, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28255571

RESUMO

The expansion of outer radial glia (oRGs, also called basal RGs) and intermediate progenitor cells (IPCs) has played a key role in the evolutionary expansion and folding of the neocortex, resulting in superior sensorimotor and cognitive abilities. In particular, oRGs, which are critical for both the increased production and lateral dispersion of neurons, are rare in lisencephalic species but vastly expanded in gyrencephalic species. However, the mechanisms that expand oRGs and IPCs are not well understood. We recently identified Sonic hedgehog (Shh) signaling as the first known signaling pathway necessary and sufficient to expand both oRGs and IPCs. Elevated Shh signaling in the embryonic neocortex leads to neocortical expansion and folding with normal cytoarchitecture in otherwise smooth mouse neocortex, whereas the loss of Shh signaling decreases oRGs, IPCs, and neocortical size. We also showed that SHH signaling activity in fetal neocortex is stronger in humans than in mice and that blocking SHH signaling decreases oRGs in human cerebral organoids. Shh signaling may be a conserved mechanism that promotes oRG and IPC expansion, driving neocortical growth and folding in humans and other species. Understanding the mechanisms underlying species-specific differences in Shh signaling activity and how Shh signaling expands oRGs and IPCs will provide insights into the mechanisms of neocortical development and evolution.

13.
Proc Natl Acad Sci U S A ; 111(34): 12438-43, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25114218

RESUMO

The apical domain of embryonic (radial glia) and adult (B1 cells) neural stem cells (NSCs) contains a primary cilium. This organelle has been suggested to function as an antenna for the detection of morphogens or growth factors. In particular, primary cilia are essential for Hedgehog (Hh) signaling, which plays key roles in brain development. Their unique location facing the ventricular lumen suggests that primary cilia in NSCs could play an important role in reception of signals within the cerebrospinal fluid. Surprisingly, ablation of primary cilia using conditional alleles for genes essential for intraflagellar transport [kinesin family member 3A (Kif3a) and intraflagellar transport 88 (Ift88)] and Cre drivers that are activated at early [Nestin; embryonic day 10.5 (E10.5)] and late [human glial fibrillary acidic protein (hGFAP); E13.5] stages of mouse neural development resulted in no apparent developmental defects. Neurogenesis in the ventricular-subventricular zone (V-SVZ) shortly after birth was also largely unaffected, except for a restricted ventral domain previously known to be regulated by Hh signaling. However, Kif3a and Ift88 genetic ablation also disrupts ependymal cilia, resulting in hydrocephalus by postnatal day 4. To directly study the role of B1 cells' primary cilia without the confounding effects of hydrocephalus, we stereotaxically targeted elimination of Kif3a from a subpopulation of radial glia, which resulted in ablation of primary cilia in a subset of B1 cells. Again, this experiment resulted in decreased neurogenesis only in the ventral V-SVZ. Primary cilia ablation led to disruption of Hh signaling in this subdomain. We conclude that primary cilia are required in a specific Hh-regulated subregion of the postnatal V-SVZ.


Assuntos
Cílios/fisiologia , Células-Tronco Neurais/classificação , Células-Tronco Neurais/ultraestrutura , Animais , Animais Recém-Nascidos , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proliferação de Células , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Feminino , Técnicas de Silenciamento de Genes , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas Hedgehog/fisiologia , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Cinesinas/metabolismo , Camundongos , Camundongos Transgênicos , Nestina/genética , Nestina/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Gravidez , Transdução de Sinais , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
14.
Mol Cell Biol ; 33(12): 2375-87, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23572559

RESUMO

The Hedgehog signaling pathway, an essential regulator of developmental patterning, has been implicated in playing causative and survival roles in a range of human cancers. The signal-transducing component of the pathway, Smoothened, has revealed itself to be an efficacious therapeutic target in combating oncogenic signaling. However, therapeutic challenges remain in cases where tumors acquire resistance to Smoothened antagonists, and also in cases where signaling is driven by active Smoothened mutants that exhibit reduced sensitivity to these compounds. We previously demonstrated that active Smoothened mutants are subjected to prolonged endoplasmic reticulum (ER) retention, likely due to their mutations triggering conformation shifts that are detected by ER quality control. We attempted to exploit this biology and demonstrate that deregulated Hedgehog signaling driven by active Smoothened mutants is specifically attenuated by ER stressors that induce the unfolded protein response (UPR). Upon UPR induction, active Smoothened mutants are targeted by ER-associated degradation, resulting in attenuation of inappropriate pathway activity. Accordingly, we found that the UPR agonist thapsigargin attenuated mutant Smoothened-induced phenotypes in vivo in Drosophila melanogaster. Wild-type Smoothened and physiological Hedgehog patterning were not affected, suggesting that UPR modulation may provide a novel therapeutic window to be evaluated for targeting active Smoothened mutants in disease.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Proteínas Hedgehog/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas/fisiologia , Células 3T3 , Animais , Antineoplásicos/farmacologia , Benzoquinonas/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Carcinoma Basocelular/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Lactamas Macrocíclicas/farmacologia , Meduloblastoma/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Pirazinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptor Smoothened , Tapsigargina/farmacologia , Ubiquitina-Proteína Ligases/genética
15.
Development ; 139(21): 3938-49, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22992951

RESUMO

The dentate gyrus of the hippocampus continues generating new neurons throughout life. These neurons originate from radial astrocytes within the subgranular zone (SGZ). Here, we find that Sox1, a member of the SoxB1 family of transcription factors, is expressed in a subset of radial astrocytes. Lineage tracing using Sox1-tTA;tetO-Cre;Rosa26 reporter mice shows that the Sox1-expressing cells represent an activated neural stem/progenitor population that gives rise to most if not all newly born granular neurons, as well as a small number of mature hilar astrocytes. Furthermore, a subpopulation of Sox1-marked cells have long-term neurogenic potential, producing new neurons 3 months after inactivation of tetracycline transactivator. Remarkably, after 8 weeks of labeling and a 12-week chase, as much as 44% of all granular neurons in the dentate gyrus were derived from Sox1 lineage-traced adult neural stem/progenitor cells. The fraction of Sox1-positive cells within the radial astrocyte population decreases with age, correlating with a decrease in neurogenesis. However, expression profiling shows that these cells are transcriptionally stable throughout the lifespan of the mouse. These results demonstrate that Sox1 is expressed in an activated stem/progenitor population whose numbers decrease with age while maintaining a stable molecular program.


Assuntos
Hipocampo/citologia , Células-Tronco Neurais/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Feminino , Citometria de Fluxo , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Camundongos
16.
Nat Cell Biol ; 12(4): 341-50, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20305650

RESUMO

In mammals, motile cilia cover many organs, such as fallopian tubes, respiratory tracts and brain ventricles. The development and function of these organs critically depend on efficient directional fluid flow ensured by the alignment of ciliary beating. To identify the mechanisms involved in this process, we analysed motile cilia of mouse brain ventricles, using biophysical and molecular approaches. Our results highlight an original orientation mechanism for ependymal cilia whereby basal bodies first dock apically with random orientations, and then reorient in a common direction through a coupling between hydrodynamic forces and the planar cell polarity (PCP) protein Vangl2, within a limited time-frame. This identifies a direct link between external hydrodynamic cues and intracellular PCP signalling. Our findings extend known PCP mechanisms by integrating hydrodynamic forces as long-range polarity signals, argue for a possible sensory role of ependymal cilia, and will be of interest for the study of fluid flow-mediated morphogenesis.


Assuntos
Polaridade Celular , Epêndima/citologia , Mecanotransdução Celular , Proteínas do Tecido Nervoso/metabolismo , Animais , Células Cultivadas , Líquido Cefalorraquidiano/metabolismo , Cílios/metabolismo , Epêndima/embriologia , Epêndima/metabolismo , Retroalimentação Fisiológica , Humanos , Cinesinas/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese , Movimento (Física) , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Recombinantes de Fusão/metabolismo , Estresse Mecânico , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/metabolismo
17.
J Neurosci ; 30(7): 2600-10, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20164345

RESUMO

Multiciliated epithelial cells, called ependymal cells, line the ventricles in the adult brain. Most ependymal cells are born prenatally and are derived from radial glia. Ependymal cells have a remarkable planar polarization that determines orientation of ciliary beating and propulsion of CSF. Disruption of ependymal ciliary beating, by injury or disease, results in aberrant CSF circulation and hydrocephalus, a common disorder of the CNS. Very little is known about the mechanisms guiding ependymal planar polarity and whether this organization is acquired during ependymal cell development or is already present in radial glia. Here we show that basal bodies in ependymal cells in the lateral ventricle walls of adult mice are polarized in two ways: (1) rotational; angle of individual basal bodies with respect to their long axis and (2) translational; the position of basal bodies on the apical surface of the cell. Conditional ablation of motile cilia disrupted rotational orientation, but translational polarity was largely preserved. In contrast, translational polarity was dramatically affected when radial glial primary cilia were ablated earlier in development. Remarkably, radial glia in the embryo have a translational polarity that predicts the orientation of mature ependymal cells. These results suggest that ependymal planar cell polarity is a multistep process initially organized by primary cilia in radial glia and then refined by motile cilia in ependymal cells.


Assuntos
Polaridade Celular/fisiologia , Epêndima/citologia , Células Epiteliais/fisiologia , Actinas/metabolismo , Animais , Cílios/fisiologia , Cílios/ultraestrutura , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Proteínas de Filamentos Intermediários/genética , Cinesinas/metabolismo , Ventrículos Laterais/citologia , Camundongos , Camundongos Transgênicos , Microscopia Confocal/métodos , Microscopia Eletrônica/métodos , Proteínas do Tecido Nervoso/genética , Nestina , Tubulina (Proteína)/metabolismo
18.
Curr Opin Neurobiol ; 20(1): 58-67, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20080044

RESUMO

The primary cilium, a hair-like extension from a cell's surface, acts as a sensory organelle to receive signals that regulate cellular behavior and physiology. Like most mammalian cells, neural progenitors and neurons have primary cilia. Recent studies show that this tiny projection plays important roles in brain development and diseases. Ciliary mutant mice show defects in brain patterning, progenitor proliferation, and specification of adult neural stem cells. Primary cilia also have dual opposing functions in the development of brain tumors. Ciliary defects are associated with genetic syndromes that frequently have neurological symptoms. Understanding the multifaceted roles that primary cilia have in brain development will provide important insights into the mechanism of brain development and diseases.


Assuntos
Encéfalo/crescimento & desenvolvimento , Neoplasias Cerebelares/etiologia , Meduloblastoma/etiologia , Neurônios/fisiologia , Animais , Padronização Corporal/fisiologia , Cílios/fisiologia , Humanos
19.
Nat Med ; 15(9): 1062-5, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19701203

RESUMO

Recent work has shown that primary cilia are essential for Hedgehog (Hh) signaling during mammalian development. It is also known that aberrant Hh signaling can lead to cancer, but the role of primary cilia in oncogenesis is not known. Cerebellar granule neuron precursors (GNPs) can give rise to medulloblastomas, the most common malignant brain tumor in children. The primary cilium and Hh signaling are required for GNP proliferation. We asked whether primary cilia in GNPs have a role in medulloblastoma growth in mice. Genetic ablation of primary cilia blocked medulloblastoma formation when this tumor was driven by a constitutively active Smoothened protein (Smo), an upstream activator of Hh signaling. In contrast, removal of cilia was required for medulloblastoma growth by a constitutively active glioma-associated oncogene family zinc finger-2 (GLI2), a downstream transcription factor. Thus, primary cilia are either required for or inhibit medulloblastoma formation, depending on the initiating oncogenic event. Remarkably, the presence or absence of cilia was associated with specific variants of human medulloblastomas; primary cilia were found in medulloblastomas with activation in HH or WNT signaling but not in most medulloblastomas in other distinct molecular subgroups. Primary cilia could serve as a diagnostic tool and provide new insights into the mechanism of tumorigenesis.


Assuntos
Neoplasias Cerebelares/etiologia , Neoplasias Cerebelares/fisiopatologia , Cílios/fisiologia , Meduloblastoma/etiologia , Meduloblastoma/fisiopatologia , Animais , Neoplasias Cerebelares/patologia , Criança , Cílios/patologia , Proteína Glial Fibrilar Ácida , Proteínas Hedgehog/fisiologia , Humanos , Cinesinas/deficiência , Cinesinas/genética , Cinesinas/fisiologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/fisiologia , Meduloblastoma/patologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais , Receptor Smoothened , Proteínas Wnt/fisiologia , Proteína Gli2 com Dedos de Zinco
20.
Cancer Cell ; 14(2): 123-34, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18691547

RESUMO

Whether the brain tumor medulloblastoma originates from stem cells or restricted progenitor cells is unclear. To investigate this, we activated oncogenic Hedgehog (Hh) signaling in multipotent and lineage-restricted central nervous system (CNS) progenitors. We observed that normal unipotent cerebellar granule neuron precursors (CGNPs) derive from hGFAP(+) and Olig2(+) rhombic lip progenitors. Hh activation in a spectrum of early- and late-stage CNS progenitors generated similar medulloblastomas, but not other brain cancers, indicating that acquisition of CGNP identity is essential for tumorigenesis. We show in human and mouse medulloblastoma that cells expressing the glia-associated markers Gfap and Olig2 are neoplastic and retain features of embryonic-type granule lineage progenitors. Thus, oncogenic Hh signaling promotes medulloblastoma from lineage-restricted granule cell progenitors.


Assuntos
Proteínas Hedgehog/metabolismo , Meduloblastoma/patologia , Neurônios/patologia , Células-Tronco/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Cérebro/patologia , Cruzamentos Genéticos , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Fenótipo , Receptores Acoplados a Proteínas G , Receptor Smoothened
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA