Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
5.
bioRxiv ; 2023 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-38014092

RESUMO

The dorsal funiculus in the spinal cord relays somatosensory information to the brain. It is made of T-shaped bifurcation of dorsal root ganglion (DRG) sensory axons. Our previous study has shown that Slit signaling is required for proper guidance during bifurcation, but loss of Slit does not affect all DRG axons. Here, we examined the role of the extracellular molecule Netrin-1 (Ntn1). Using wholemount staining with tissue clearing, we showed that mice lacking Ntn1 have axons escaping from the dorsal funiculus at the time of bifurcation. Genetic labeling confirmed that these misprojecting axons come from DRG neurons. Single axon analysis showed that the defect does not affect bifurcation but rather alters turning angles. To distinguish their guidance functions, we examined mice with triple deletion of Ntn1, Slit2, and Slit2 and found a completely disorganized dorsal funiculus. Comparing mice with different genotypes using immunolabeling and single axon tracing revealed additive guidance defects, demonstrating the independent roles of Ntn1 and Slit. Moreover, the same defects were observed in embryos lacking their cognate receptors. These in vivo studies thus demonstrate the presence of multi-factorial guidance mechanisms that ensure proper formation of a common branched axonal structure during spinal cord development.

8.
Life Sci Alliance ; 6(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37311584

RESUMO

SLIT/ROBO signaling impacts many aspects of tissue development and homeostasis, in part, through the regulation of cell growth and proliferation. Recent studies have also linked SLIT/ROBO signaling to the regulation of diverse phagocyte functions. However, the mechanisms by which SLIT/ROBO signaling acts at the nexus of cellular growth control and innate immunity remain enigmatic. Here, we show that SLIT2-mediated activation of ROBO1 leads to inhibition of mTORC1 kinase activity in macrophages, leading to dephosphorylation of its downstream targets, including transcription factor EB and ULK1. Consequently, SLIT2 augments lysosome biogenesis, potently induces autophagy, and robustly promotes the killing of bacteria within phagosomes. Concordant with these results, we demonstrate decreased lysosomal content and accumulated peroxisomes in the spinal cords of embryos from Robo1 -/- , Robo2 -/- double knockout mice. We also show that impediment of auto/paracrine SLIT-ROBO signaling axis in cancer cells leads to hyperactivation of mTORC1 and inhibition of autophagy. Together, these findings elucidate a central role of chemorepellent SLIT2 in the regulation of mTORC1 activity with important implications for innate immunity and cancer cell survival.


Assuntos
Proteínas do Tecido Nervoso , Receptores Imunológicos , Animais , Camundongos , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Lisossomos , Bactérias , Alvo Mecanístico do Complexo 1 de Rapamicina
12.
EMBO J ; 42(1): e110565, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36377476

RESUMO

Cortical neuronal networks control cognitive output, but their composition and modulation remain elusive. Here, we studied the morphological and transcriptional diversity of cortical cholinergic VIP/ChAT interneurons (VChIs), a sparse population with a largely unknown function. We focused on VChIs from the whole barrel cortex and developed a high-throughput automated reconstruction framework, termed PopRec, to characterize hundreds of VChIs from each mouse in an unbiased manner, while preserving 3D cortical coordinates in multiple cleared mouse brains, accumulating thousands of cells. We identified two fundamentally distinct morphological types of VChIs, bipolar and multipolar that differ in their cortical distribution and general morphological features. Following mild unilateral whisker deprivation on postnatal day seven, we found after three weeks both ipsi- and contralateral dendritic arborization differences and modified cortical depth and distribution patterns in the barrel fields alone. To seek the transcriptomic drivers, we developed NuNeX, a method for isolating nuclei from fixed tissues, to explore sorted VChIs. This highlighted differentially expressed neuronal structural transcripts, altered exitatory innervation pathways and established Elmo1 as a key regulator of morphology following deprivation.


Assuntos
Lobo Parietal , Transcriptoma , Camundongos , Animais , Interneurônios/fisiologia , Colina O-Acetiltransferase , Colinérgicos/metabolismo , Células Receptoras Sensoriais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
13.
Neuron ; 110(22): 3656-3660, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36356578

RESUMO

In May, an interdisciplinary group gathered in Crete for the Molecular Neurobiology Workshop. Scientists shared data acquired by vastly diverse techniques to understand how the nervous system, with only a limited number of components, is assembled to respond to infinite stimuli. Ideas of molecular cues, timing, switching, and context emerged.


Assuntos
Neurociências , Neurobiologia
15.
Proc Natl Acad Sci U S A ; 119(43): e2210122119, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36256819

RESUMO

Hyperexcitability of brain circuits is a common feature of autism spectrum disorders (ASDs). Genetic deletion of a chromatin-binding protein, retinoic acid induced 1 (RAI1), causes Smith-Magenis syndrome (SMS). SMS is a syndromic ASD associated with intellectual disability, autistic features, maladaptive behaviors, overt seizures, and abnormal electroencephalogram (EEG) patterns. The molecular and neural mechanisms underlying abnormal brain activity in SMS remain unclear. Here we show that panneural Rai1 deletions in mice result in increased seizure susceptibility and prolonged hippocampal seizure duration in vivo and increased dentate gyrus population spikes ex vivo. Brain-wide mapping of neuronal activity pinpointed selective cell types within the limbic system, including the hippocampal dentate gyrus granule cells (dGCs) that are hyperactivated by chemoconvulsant administration or sensory experience in Rai1-deficient brains. Deletion of Rai1 from glutamatergic neurons, but not from gamma-aminobutyric acidergic (GABAergic) neurons, was responsible for increased seizure susceptibility. Deleting Rai1 from the Emx1Cre-lineage glutamatergic neurons resulted in abnormal dGC properties, including increased excitatory synaptic transmission and increased intrinsic excitability. Our work uncovers the mechanism of neuronal hyperexcitability in SMS by identifying Rai1 as a negative regulator of dGC intrinsic and synaptic excitability.


Assuntos
Síndrome de Smith-Magenis , Camundongos , Animais , Síndrome de Smith-Magenis/genética , Transativadores/genética , Transativadores/metabolismo , Fenótipo , Modelos Animais de Doenças , Cromatina , Hipocampo/metabolismo , Convulsões/genética , Tretinoína
16.
Dev Cell ; 56(13): 1900-1916.e5, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34197726

RESUMO

Stem cells (SCs) play a key role in homeostasis and repair. While many studies have focused on SC self-renewal and differentiation, little is known regarding the molecular mechanism regulating SC elimination and compensation upon loss. Here, we report that Caspase-9 deletion in hair follicle SCs (HFSCs) attenuates the apoptotic cascade, resulting in significant temporal delays. Surprisingly, Casp9-deficient HFSCs accumulate high levels of cleaved caspase-3 and are improperly cleared due to an essential caspase-3/caspase-9 feedforward loop. These SCs are retained in an apoptotic-engaged state, serving as mitogenic signaling centers by continuously releasing Wnt3 and instructing proliferation. Investigating the underlying mechanism, we reveal a caspase-3/Dusp8/p38 module responsible for Wnt3 induction, which operates in both normal and Casp9-deleted HFSCs. Notably, Casp9-deleted mice display accelerated wound repair and de novo hair follicle regeneration. Taken together, we demonstrate that apoptotic cells represent a dynamic SC niche, from which emanating signals drive SC proliferation and tissue regeneration.


Assuntos
Caspase 3/genética , Caspase 9/genética , Fosfatases de Especificidade Dupla/genética , Regeneração/genética , Proteína Wnt3/genética , Animais , Apoptose/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular/genética , Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Nicho de Células-Tronco/genética , Células-Tronco/metabolismo , Cicatrização/genética
17.
Dev Cell ; 56(7): 976-984.e3, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33823136

RESUMO

Axon remodeling through sprouting and pruning contributes to the refinement of developing neural circuits. A prominent example is the pruning of developing sensory axons deprived of neurotrophic support, which is mediated by a caspase-dependent (apoptotic) degeneration process. Distal sensory axons possess a latent apoptotic pathway, but a cell body-derived signal that travels anterogradely down the axon is required for pathway activation. The signaling mechanisms that underlie this anterograde process are poorly understood. Here, we show that the tumor suppressor P53 is required for anterograde signaling. Interestingly loss of P53 blocks axonal but not somatic (i.e., cell body) caspase activation. Unexpectedly, P53 does not appear to have an acute transcriptional role in this process and instead appears to act in the cytoplasm to directly activate the mitochondrial apoptotic pathway in axons. Our data support the operation of a cytoplasmic role for P53 in the anterograde death of developing sensory axons.


Assuntos
Axônios/fisiologia , Células Receptoras Sensoriais/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Axônios/enzimologia , Axônios/metabolismo , Caspases/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Camundongos , Domínios Proteicos , Células Receptoras Sensoriais/enzimologia , Células Receptoras Sensoriais/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteína bcl-X/antagonistas & inibidores
18.
Cell ; 184(3): 689-708.e20, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33482083

RESUMO

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.


Assuntos
Proteína C9orf72/metabolismo , Expansão das Repetições de DNA/genética , Degeneração Neural/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Axônios/metabolismo , Proteína C9orf72/genética , Morte Celular , Células Cultivadas , Córtex Cerebral/patologia , Cromatina/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Drosophila , Camundongos Endogâmicos C57BL , Degeneração Neural/patologia , Estabilidade Proteica , Transcrição Gênica , Proteínas Supressoras de Tumor/metabolismo
19.
Neuron ; 107(6): 1113-1123.e4, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32679036

RESUMO

Disrupting memories that associate environmental cues with drug experiences holds promise for treating addiction, yet accessing the distributed neural network that stores such memories is challenging. Here, we show that the paraventricular nucleus of the thalamus (PVT) orchestrates the acquisition and maintenance of opiate-associated memories via projections to the central nucleus of the amygdala (CeA) and nucleus accumbens (NAc). PVT→CeA activity associates morphine reward to the environment, whereas transient inhibition of the PVT→NAc pathway during retrieval causes enduring protection against opiate-primed relapse. Using brain-wide activity mapping, we revealed distributed network activities that are altered in non-relapsing mice, which enabled us to find that activating the downstream NAc→lateral hypothalamus (LH) pathway also prevents relapse. These findings establish the PVT as a key node in the opiate-associated memory network and demonstrate the potential of targeting the PVT→NAc→LH pathway for treating opioid addiction.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Núcleo Accumbens/fisiopatologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Priming de Repetição , Animais , Sinais (Psicologia) , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/fisiopatologia
20.
J Neurosci ; 40(28): 5402-5412, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32471877

RESUMO

Axon guidance molecules and neuronal activity have been implicated in the establishment and refinement of neural circuits during development. It is unclear, however, whether these guidance molecule- and activity-dependent mechanisms interact with one another to shape neural circuit formation. The formation of corticospinal (CS) circuits, which are essential for voluntary movements, involves both guidance molecule- and activity-dependent components during development. We previously showed that semaphorin6D (Sema6D)-plexinA1 (PlexA1) signaling eliminates ipsilateral projections of CS neurons in the spinal cord, while other studies demonstrate that CS projections to the spinal cord are eliminated in an activity-dependent manner. Here we show that inhibition of cortical neurons during postnatal development causes defects in elimination of ipsilateral CS projections in mice. We further show that mice that lack the activity-dependent Bax/Bak pathway or caspase-9 similarly exhibit defects in elimination of ipsilateral CS projections, suggesting that the activity-dependent Bax/Bak-caspase-9 pathway is essential for the removal of ipsilateral CS projections. Interestingly, either inhibition of neuronal activity in the cortex or deletion of Bax/Bak in mice causes a reduction in PlexA1 protein expression in corticospinal neurons. Finally, intracortical microstimulation induces activation of only contralateral forelimb muscles in control mice, whereas it induces activation of both contralateral and ipsilateral muscles in mice with cortical inhibition, suggesting that the ipsilaterally projecting CS axons that have been maintained in mice with cortical inhibition form functional connections. Together, these results provide evidence of a potential link between the repellent signaling of Sema6D-PlexA1 and neuronal activity to regulate axon elimination.SIGNIFICANCE STATEMENT Both axon guidance molecules and neuronal activity regulate axon elimination to refine neuronal circuits during development. However, the degree to which these mechanisms operate independently or cooperatively to guide network generation is unclear. Here, we show that neuronal activity-driven Bax/Bak-caspase signaling induces expression of the PlexA1 receptor for the repellent Sema6D molecule in corticospinal neurons (CSNs). This cascade eliminates ipsilateral projections of CSNs in the spinal cord during early postnatal development. The absence of PlexA1, neuronal activity, Bax and Bak, or caspase-9 leads to the maintenance of ipsilateral projections of CSNs, which can form functional connections with spinal neurons. Together, these studies reveal how the Sema6D-PlexA1 signaling and neuronal activity may play a cooperative role in refining CS axonal projections.


Assuntos
Axônios/metabolismo , Caspases/metabolismo , Tratos Piramidais/crescimento & desenvolvimento , Semaforinas/metabolismo , Transdução de Sinais/fisiologia , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Orientação de Axônios/fisiologia , Camundongos , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Neurônios/fisiologia , Tratos Piramidais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...