Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.776
Filtrar
1.
J Cell Biol ; 223(8)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38713825

RESUMO

Whether, to what extent, and how the axons in the central nervous system (CNS) can withstand sudden mechanical impacts remain unclear. By using a microfluidic device to apply controlled transverse mechanical stress to axons, we determined the stress levels that most axons can withstand and explored their instant responses at nanoscale resolution. We found mild stress triggers a highly reversible, rapid axon beading response, driven by actomyosin-II-dependent dynamic diameter modulations. This mechanism contributes to hindering the long-range spread of stress-induced Ca2+ elevations into non-stressed neuronal regions. Through pharmacological and molecular manipulations in vitro, we found that actomyosin-II inactivation diminishes the reversible beading process, fostering progressive Ca2+ spreading and thereby increasing acute axonal degeneration in stressed axons. Conversely, upregulating actomyosin-II activity prevents the progression of initial injury, protecting stressed axons from acute degeneration both in vitro and in vivo. Our study unveils the periodic actomyosin-II in axon shafts cortex as a novel protective mechanism, shielding neurons from detrimental effects caused by mechanical stress.


Assuntos
Actomiosina , Axônios , Estresse Mecânico , Animais , Camundongos , Actomiosina/metabolismo , Axônios/metabolismo , Axônios/patologia , Cálcio/metabolismo , Células Cultivadas , Degeneração Neural/patologia , Ratos
2.
PLoS Genet ; 20(5): e1011253, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38722918

RESUMO

Synaptic vesicle proteins (SVps) are transported by the motor UNC-104/KIF1A. We show that SVps travel in heterogeneous carriers in C. elegans neuronal processes, with some SVp carriers co-transporting lysosomal proteins (SV-lysosomes). LRK-1/LRRK2 and the clathrin adaptor protein complex AP-3 play a critical role in the sorting of SVps and lysosomal proteins away from each other at the SV-lysosomal intermediate trafficking compartment. Both SVp carriers lacking lysosomal proteins and SV-lysosomes are dependent on the motor UNC-104/KIF1A for their transport. In lrk-1 mutants, both SVp carriers and SV-lysosomes can travel in axons in the absence of UNC-104, suggesting that LRK-1 plays an important role to enable UNC-104 dependent transport of synaptic vesicle proteins. Additionally, LRK-1 acts upstream of the AP-3 complex and regulates its membrane localization. In the absence of the AP-3 complex, the SV-lysosomes become more dependent on the UNC-104-SYD-2/Liprin-α complex for their transport. Therefore, SYD-2 acts to link upstream trafficking events with the transport of SVps likely through its interaction with the motor UNC-104. We further show that the mistrafficking of SVps into the dendrite in lrk-1 and apb-3 mutants depends on SYD-2, likely by regulating the recruitment of the AP-1/UNC-101. SYD-2 acts in concert with AP complexes to ensure polarized trafficking & transport of SVps.


Assuntos
Complexo 3 de Proteínas Adaptadoras , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Lisossomos , Proteínas do Tecido Nervoso , Vesículas Sinápticas , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/genética , Complexo 3 de Proteínas Adaptadoras/metabolismo , Complexo 3 de Proteínas Adaptadoras/genética , Lisossomos/metabolismo , Lisossomos/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Transporte Proteico , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Neurônios/metabolismo , Cinesinas/metabolismo , Cinesinas/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Axônios/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular
3.
Biofabrication ; 16(3)2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38565133

RESUMO

Spinal cord injury (SCI) can cause permanent impairment to motor or sensory functions. Pre-cultured neural stem cell (NSC) hydrogel scaffolds have emerged as a promising approach to treat SCI by promoting anti-inflammatory effects, axon regrowth, and motor function restoration. Here, in this study, we performed a coaxial extrusion process to fabricate a core-shell hydrogel microfiber with high NSC density in the core portion. Oxidized hyaluronic acid, carboxymethyl chitosan, and matrigel blend were used as a matrix for NSC growth and to facilitate the fabrication process. During thein vitrodifferentiation culture, it was found that NSC microfibers could differentiate into neurons and astrocytes with higher efficiency compared to NSC cultured in petri dishes. Furthermore, duringin vivotransplantation, NSC microfibers were coated with polylactic acid nanosheets by electrospinning for reinforcement. The coated NSC nanofibers exhibited higher anti-inflammatory effect and lesion cavity filling rate compared with the control group. Meanwhile, more neuron- and oligodendrocyte-like cells were visualized at the lesion epicenter. Finally, axon regrowth across the whole lesion site was observed, demonstrating that the microfiber could guide renascent axon regrowth. Experiment results indicate that the NSC microfiber is a promising bioactive treatment for complete SCI treatment with superior outcomes.


Assuntos
Axônios , Diferenciação Celular , Células-Tronco Neurais , Neurônios , Traumatismos da Medula Espinal , Alicerces Teciduais , Animais , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/patologia , Axônios/efeitos dos fármacos , Axônios/fisiologia , Axônios/metabolismo , Diferenciação Celular/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Alicerces Teciduais/química , Ratos Sprague-Dawley , Hidrogéis/química , Hidrogéis/farmacologia , Quitosana/química , Quitosana/farmacologia , Quitosana/análogos & derivados , Células Cultivadas , Regeneração Nervosa/efeitos dos fármacos , Nanofibras/química , Ratos , Feminino
4.
Amino Acids ; 56(1): 32, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38637413

RESUMO

Diabetic neuropathy (DN) is a common neurological complication caused by diabetes mellitus (DM). Axonal degeneration is generally accepted to be the major pathological change in peripheral DN. Taurine has been evidenced to be neuroprotective in various aspects, but its effect on spinal cord axon injury (SCAI) in DN remains barely reported. This study showed that taurine significantly ameliorated axonal damage of spinal cord (SC), based on morphological and functional analyses, in a rat model of DN induced by streptozotocin (STZ). Taurine was also found to induce neurite outgrowth in cultured cerebral cortex neurons with high glucose exposure. Moreover, taurine up-regulated the expression of nerve growth factor (NGF) and neurite outgrowth relative protein GAP-43 in rat DN model and cultured cortical neurons/VSC4.1 cells. Besides, taurine increased the activating phosphorylation signals of TrkA, Akt, and mTOR. Mechanistically, the neuroprotection by taurine was related to the NGF-pAKT-mTOR axis, because either NGF-neutralizing antibody or Akt or mTOR inhibitors was found to attenuate its beneficial effects. Together, our results demonstrated that taurine promotes spinal cord axon repair in a model of SCAI in STZ-induced diabetic rats, mechanistically associating with the NGF-dependent activation of Akt/mTOR pathway.


Assuntos
Diabetes Mellitus Experimental , Proteínas Proto-Oncogênicas c-akt , Animais , Ratos , Axônios/metabolismo , Axônios/patologia , Diabetes Mellitus Experimental/metabolismo , Fator de Crescimento Neural/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Taurina/farmacologia , Taurina/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
5.
Curr Biol ; 34(9): 1904-1917.e6, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38642548

RESUMO

Neurons have differential and fluctuating energy needs across distinct cellular compartments, shaped by brain electrochemical activity associated with cognition. In vitro studies show that mitochondria transport from soma to axons is key to maintaining neuronal energy homeostasis. Nevertheless, whether the spatial distribution of neuronal mitochondria is dynamically adjusted in vivo in an experience-dependent manner remains unknown. In Drosophila, associative long-term memory (LTM) formation is initiated by an early and persistent upregulation of mitochondrial pyruvate flux in the axonal compartment of neurons in the mushroom body (MB). Through behavior experiments, super-resolution analysis of mitochondria morphology in the neuronal soma and in vivo mitochondrial fluorescence recovery after photobleaching (FRAP) measurements in the axons, we show that LTM induction, contrary to shorter-lived memories, is sustained by the departure of some mitochondria from MB neuronal soma and increased mitochondrial dynamics in the axonal compartment. Accordingly, impairing mitochondrial dynamics abolished the increased pyruvate consumption, specifically after spaced training and in the MB axonal compartment, thereby preventing LTM formation. Our results thus promote reorganization of the mitochondrial network in neurons as an integral step in elaborating high-order cognitive processes.


Assuntos
Axônios , Proteínas de Drosophila , Drosophila melanogaster , Memória de Longo Prazo , Mitocôndrias , Dinâmica Mitocondrial , Corpos Pedunculados , Animais , Memória de Longo Prazo/fisiologia , Dinâmica Mitocondrial/fisiologia , Axônios/metabolismo , Axônios/fisiologia , Corpos Pedunculados/fisiologia , Corpos Pedunculados/metabolismo , Drosophila melanogaster/fisiologia , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Neurônios/metabolismo , Neurônios/fisiologia
6.
Cell Commun Signal ; 22(1): 236, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38650003

RESUMO

BACKGROUND: The preservation of retinal ganglion cells (RGCs) and the facilitation of axon regeneration are crucial considerations in the management of various vision-threatening disorders. Therefore, we investigate the efficacy of interleukin-4 (IL-4), a potential therapeutic agent, in promoting neuroprotection and axon regeneration of retinal ganglion cells (RGCs) as identified through whole transcriptome sequencing in an in vitro axon growth model. METHODS: A low concentration of staurosporine (STS) was employed to induce in vitro axon growth. Whole transcriptome sequencing was utilized to identify key target factors involved in the molecular mechanism underlying axon growth. The efficacy of recombinant IL-4 protein on promoting RGC axon growth was validated through in vitro experiments. The protective effect of recombinant IL-4 protein on somas of RGCs was assessed using RBPMS-specific immunofluorescent staining in mouse models with optic nerve crush (ONC) and N-methyl-D-aspartic acid (NMDA) injury. The protective effect on RGC axons was evaluated by anterograde labeling of cholera toxin subunit B (CTB), while the promotion of RGC axon regeneration was assessed through both anterograde labeling of CTB and immunofluorescent staining for growth associated protein-43 (GAP43). RESULTS: Whole-transcriptome sequencing of staurosporine-treated 661 W cells revealed a significant upregulation in intracellular IL-4 transcription levels during the process of axon regeneration. In vitro experiments demonstrated that recombinant IL-4 protein effectively stimulated axon outgrowth. Subsequent immunostaining with RBPMS revealed a significantly higher survival rate of RGCs in the rIL-4 group compared to the vehicle group in both NMDA and ONC injury models. Axonal tracing with CTB confirmed that recombinant IL-4 protein preserved long-distance projection of RGC axons, and there was a notably higher number of surviving axons in the rIL-4 group compared to the vehicle group following NMDA-induced injury. Moreover, intravitreal delivery of recombinant IL-4 protein substantially facilitated RGC axon regeneration after ONC injury. CONCLUSION: The recombinant IL-4 protein exhibits the potential to enhance the survival rate of RGCs, protect RGC axons against NMDA-induced injury, and facilitate axon regeneration following ONC. This study provides an experimental foundation for further investigation and development of therapeutic agents aimed at protecting the optic nerve and promoting axon regeneration.


Assuntos
Axônios , Interleucina-4 , Regeneração Nervosa , Células Ganglionares da Retina , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/metabolismo , Animais , Interleucina-4/farmacologia , Axônios/efeitos dos fármacos , Axônios/metabolismo , Regeneração Nervosa/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/tratamento farmacológico , N-Metilaspartato/farmacologia , Estaurosporina/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteínas Recombinantes/farmacologia
7.
Genesis ; 62(1): e23586, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38593162

RESUMO

Neural activity influences every aspect of nervous system development. In olfactory systems, sensory neurons expressing the same odorant receptor project their axons to stereotypically positioned glomeruli, forming a spatial map of odorant receptors in the olfactory bulb. As individual odors activate unique combinations of glomeruli, this map forms the basis for encoding olfactory information. The establishment of this stereotypical olfactory map requires coordinated regulation of axon guidance molecules instructed by spontaneous activity. Recent studies show that sensory experiences also modify innervation patterns in the olfactory bulb, especially during a critical period of the olfactory system development. This review examines evidence in the field to suggest potential mechanisms by which various aspects of neural activity regulate axon targeting. We also discuss the precise functions served by neural plasticity during the critical period.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Neurônios Receptores Olfatórios/metabolismo , Bulbo Olfatório/fisiologia , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Axônios/metabolismo , Mamíferos
8.
PLoS One ; 19(4): e0302251, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38635746

RESUMO

Sterile alpha and TIR motif-containing 1 (SARM1) is a protein involved in programmed death of injured axons. Following axon injury or a drug-induced insult, the TIR domain of SARM1 degrades the essential molecule nicotinamide adenine dinucleotide (NAD+), leading to a form of axonal death called Wallerian degeneration. Degradation of NAD+ by SARM1 is essential for the Wallerian degeneration process, but accumulating evidence suggest that other activities of SARM1, beyond the mere degradation of NAD+, may be necessary for programmed axonal death. In this study we show that the TIR domains of both human and fruit fly SARM1 produce 1''-2' and 1''-3' glycocyclic ADP-ribose (gcADPR) molecules as minor products. As previously reported, we observed that SARM1 TIR domains mostly convert NAD+ to ADPR (for human SARM1) or cADPR (in the case of SARM1 from Drosophila melanogaster). However, we now show that human and Drosophila SARM1 additionally convert ~0.1-0.5% of NAD+ into gcADPR molecules. We find that SARM1 TIR domains produce gcADPR molecules both when purified in vitro and when expressed in bacterial cells. Given that gcADPR is a second messenger involved in programmed cell death in bacteria and likely in plants, we propose that gcADPR may play a role in SARM1-induced programmed axonal death in animals.


Assuntos
NAD , Degeneração Walleriana , Animais , Humanos , Degeneração Walleriana/metabolismo , Degeneração Walleriana/patologia , NAD/metabolismo , Drosophila melanogaster/metabolismo , Axônios/metabolismo , Bactérias/metabolismo , Adenosina Difosfato Ribose/metabolismo , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo
9.
Phys Rev E ; 109(3-1): 034401, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38632795

RESUMO

The diffusive ion current is insufficient to explain the fast saltatory conduction observed in myelinated axons and in pain-sensing C fibers in the human nervous system, where the stimulus signal exhibits a velocity two orders of magnitude greater than the upper limit of ion diffusion velocity, even when the diffusion is accelerated by myelin, as in the discrete cable model including the Hodgkin-Huxley mechanism. The agreement with observations has been achieved in a wave-type model of stimulus signal kinetics via synchronized ion local density oscillations propagating as a wave in axons periodically corrugated by myelin segments in myelinated axons, or by periodically distributed rafts with clusters of Na^{+} channels in C fibers. The resulting so-called plasmon-polariton model for saltatory conduction reveals also the specific role of myelin, which is different from what was previously thought. This can be important for identifying a new target for the future treatment of demyelination diseases.


Assuntos
Bainha de Mielina , Condução Nervosa , Humanos , Condução Nervosa/fisiologia , Bainha de Mielina/fisiologia , Axônios/metabolismo , Transporte de Íons , Simulação por Computador , Potenciais de Ação/fisiologia
10.
PLoS One ; 19(4): e0300539, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38574058

RESUMO

Genetic and pharmacological perturbation of the cytoskeleton enhances the regenerative potential of neurons. This response requires Dual-leucine Zipper Kinase (DLK), a neuronal stress sensor that is a central regulator of axon regeneration and degeneration. The damage and repair aspects of this response are reminiscent of other cellular homeostatic systems, suggesting that a cytoskeletal homeostatic response exists. In this study, we propose a framework for understanding DLK mediated neuronal cytoskeletal homeostasis. We demonstrate that low dose nocodazole treatment activates DLK signaling. Activation of DLK signaling results in a DLK-dependent transcriptional signature, which we identify through RNA-seq. This signature includes genes likely to attenuate DLK signaling while simultaneously inducing actin regulating genes. We identify alterations to the cytoskeleton including actin-based morphological changes to the axon. These results are consistent with the model that cytoskeletal disruption in the neuron induces a DLK-dependent homeostatic mechanism, which we term the Cytoskeletal Stress Response (CSR) pathway.


Assuntos
Actinas , Axônios , Axônios/metabolismo , Nocodazol/farmacologia , Actinas/metabolismo , Zíper de Leucina , Regeneração Nervosa/fisiologia , Citoesqueleto/metabolismo , Homeostase , MAP Quinase Quinase Quinases/genética
11.
Int J Mol Sci ; 25(8)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38673871

RESUMO

Mild traumatic brain injury (mTBI) affects millions of people in the U.S. Approximately 20-30% of those individuals develop adverse symptoms lasting at least 3 months. In a rat mTBI study, the closed-head impact model of engineered rotational acceleration (CHIMERA) produced significant axonal injury in the optic tract (OT), indicating white-matter damage. Because retinal ganglion cells project to the lateral geniculate nucleus (LGN) in the thalamus through the OT, we hypothesized that synaptic density may be reduced in the LGN of rats following CHIMERA injury. A modified SEQUIN (synaptic evaluation and quantification by imaging nanostructure) method, combined with immunofluorescent double-labeling of pre-synaptic (synapsin) and post-synaptic (PSD-95) markers, was used to quantify synaptic density in the LGN. Microglial activation at the CHIMERA injury site was determined using Iba-1 immunohistochemistry. Additionally, the effects of ketamine, a potential neuroprotective drug, were evaluated in CHIMERA-induced mTBI. A single-session repetitive (ssr-) CHIMERA (3 impacts, 1.5 joule/impact) produced mild effects on microglial activation at the injury site, which was significantly enhanced by post-injury intravenous ketamine (10 mg/kg) infusion. However, ssr-CHIMERA did not alter synaptic density in the LGN, although ketamine produced a trend of reduction in synaptic density at post-injury day 4. Further research is necessary to characterize the effects of ssr-CHIMERA and subanesthetic doses of intravenous ketamine on different brain regions and multiple time points post-injury. The current study demonstrates the utility of the ssr-CHIMERA as a rodent model of mTBI, which researchers can use to identify biological mechanisms of mTBI and to develop improved treatment strategies for individuals suffering from head trauma.


Assuntos
Ketamina , Microglia , Ratos Sprague-Dawley , Sinapses , Animais , Ketamina/administração & dosagem , Ketamina/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Ratos , Masculino , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia , Traumatismos Cranianos Fechados/patologia , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Modelos Animais de Doenças , Corpos Geniculados/patologia , Corpos Geniculados/efeitos dos fármacos , Concussão Encefálica/patologia , Concussão Encefálica/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Sinapsinas/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/administração & dosagem
12.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664711

RESUMO

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Assuntos
Fator 3 Ativador da Transcrição , Axônios , Quebras de DNA de Cadeia Dupla , Gânglios Espinais , Células-Tronco Mesenquimais , Mitocôndrias , Regeneração Nervosa , Espécies Reativas de Oxigênio , Nervo Isquiático , Regulação para Cima , Animais , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/genética , Espécies Reativas de Oxigênio/metabolismo , Axônios/metabolismo , Regeneração Nervosa/genética , Regulação para Cima/genética , Camundongos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Gânglios Espinais/metabolismo , Camundongos Endogâmicos C57BL , Masculino
13.
PLoS Genet ; 20(4): e1011139, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38669217

RESUMO

As essential components of gene expression networks, transcription factors regulate neural circuit assembly. The homeobox transcription factor encoding gene, gs homeobox 1 (gsx1), is expressed in the developing visual system; however, no studies have examined its role in visual system formation. In zebrafish, retinal ganglion cell (RGC) axons that transmit visual information to the brain terminate in ten arborization fields (AFs) in the optic tectum (TeO), pretectum (Pr), and thalamus. Pretectal AFs (AF1-AF9) mediate distinct visual behaviors, yet we understand less about their development compared to AF10 in the TeO. Using gsx1 zebrafish mutants, immunohistochemistry, and transgenic lines, we observed that gsx1 is required for vesicular glutamate transporter, Tg(slc17a6b:DsRed), expression in the Pr, but not overall neuron number. gsx1 mutants have normal eye morphology, yet they exhibit impaired visual ability during prey capture. RGC axon volume in the gsx1 mutant Pr and TeO is reduced, and AF7 that is active during feeding is missing which is consistent with reduced hunting performance. Timed laser ablation of Tg(slc17a6b:DsRed)-positive cells reveals that they are necessary for AF7 formation. This work is the first to implicate gsx1 in establishing cell identity and functional neural circuits in the visual system.


Assuntos
Animais Geneticamente Modificados , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio , Células Ganglionares da Retina , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Axônios/metabolismo , Axônios/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Mutação , Células Ganglionares da Retina/metabolismo , Colículos Superiores/metabolismo , Colículos Superiores/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vias Visuais/crescimento & desenvolvimento , Vias Visuais/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
14.
Genesis ; 62(2): e23594, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38590146

RESUMO

During development of the nervous system, neurons connect to one another in a precisely organized manner. Sensory systems provide a good example of this organization, whereby the composition of the outside world is represented in the brain by neuronal maps. Establishing correct patterns of neural circuitry is crucial, as inaccurate map formation can lead to severe disruptions in sensory processing. In rodents, olfactory stimuli modulate a wide variety of behaviors essential for survival. The formation of the olfactory glomerular map is dependent on molecular cues that guide olfactory receptor neuron axons to broad regions of the olfactory bulb and on cell adhesion molecules that promote axonal sorting into specific synaptic units in this structure. Here, we demonstrate that the cell adhesion molecule Amigo1 is expressed in a subpopulation of olfactory receptor neurons, and we investigate its role in the precise targeting of olfactory receptor neuron axons to the olfactory bulb using a genetic loss-of-function approach in mice. While ablation of Amigo1 did not lead to alterations in olfactory sensory neuron axonal targeting, our experiments revealed that the presence of a neomycin resistance selection cassette in the Amigo1 locus can lead to off-target effects that are not due to loss of Amigo1 expression, including unexpected altered gene expression in olfactory receptor neurons and reduced glomerular size in the ventral region of the olfactory bulb. Our results demonstrate that insertion of a neomycin selection cassette into the mouse genome can have specific deleterious effects on the development of the olfactory system and highlight the importance of removing antibiotic resistance cassettes from genetic loss-of-function mouse models when studying olfactory system development.


Assuntos
Neurônios Receptores Olfatórios , Animais , Camundongos , Neurônios Receptores Olfatórios/metabolismo , Mucosa Olfatória , Bulbo Olfatório , Axônios/metabolismo , Expressão Gênica
15.
Acta Neurobiol Exp (Wars) ; 84(1): 80-88, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38587321

RESUMO

Diffuse axonal injury (DAI), one of the most common and devastating type of traumatic brain injury, is the result of the shear force on axons due to severe rotational acceleration and deceleration. Neurogranin (NRGN) is a postsynaptic protein secreted by excitatory neurons, and synaptic dysfunction can alter extracellular NRGN levels. In this study, we examined NRGN levels in serum and cerebrospinal fluid (CSF) after experimental DAI in terms of their diagnostic value. Experimental DAI was induced using the Marmarou technique in male Wistar albino rats. Serum and CSF NRGN levels of the sham group, one­hour, six­hour, 24­hour, and 72­hour post­DAI groups were measured by ELISA method. DAI was verified by staining with hematoxylin­eosin and ß­amyloid precursor protein in the rat brain samples. While no histopathological and immunohistochemical changes were observed in the early hours of the post­DAI groups, the staining of the ß­APP visibly increased over time, with positivity being most frequent and intense in the 72­hour group. It was found that serum NRGN levels were significantly lower in the 6­hour group than in the sham group. The serum NRGN levels in the 24­hour group were significantly higher than those in the sham group. This study showed a dichotomy of post­DAI serum NRGN levels in consecutive time periods. NRGN levels in CSF were higher in the one­hour group than in the sham group and returned to baseline by 72 hours, although not significantly. Our study provides an impression of serum and CSF NRGN levels in a rat DAI model in consecutive time periods. Further studies are needed to understand the diagnostic value of NRGN.


Assuntos
Lesão Axonal Difusa , Neurogranina , Ratos , Masculino , Animais , Neurogranina/metabolismo , Ratos Wistar , Lesão Axonal Difusa/metabolismo , Lesão Axonal Difusa/patologia , Neurônios/metabolismo , Axônios/metabolismo
16.
Biochem Biophys Res Commun ; 712-713: 149932, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38626530

RESUMO

The DHCR7 enzyme converts 7-DHC into cholesterol. Mutations in DHCR7 can block cholesterol production, leading to abnormal accumulation of 7-DHC and causing Smith-Lemli-Opitz syndrome (SLOS). SLOS is an autosomal recessive disorder characterized by multiple malformations, including microcephaly, intellectual disability, behavior reminiscent of autism, sleep disturbances, and attention-deficit/hyperactivity disorder (ADHD)-like hyperactivity. Although 7-DHC affects neuronal differentiation in ex vivo experiments, the precise mechanism of SLOS remains unclear. We generated Dhcr7 deficient (dhcr7-/-) zebrafish that exhibited key features of SLOS, including microcephaly, decreased neural stem cell pools, and behavioral phenotypes similar to those of ADHD-like hyperactivity. These zebrafish demonstrated compromised myelination, synaptic anomalies, and neurotransmitter imbalances. The axons of the dhcr7-/- zebrafish showed increased lysosomes and attenuated autophagy, suggesting that autophagy-related neuronal homeostasis is disrupted.


Assuntos
Axônios , Colesterol , Síndrome de Smith-Lemli-Opitz , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Peixe-Zebra/genética , Colesterol/metabolismo , Axônios/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Síndrome de Smith-Lemli-Opitz/genética , Síndrome de Smith-Lemli-Opitz/patologia , Neurogênese , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/deficiência , Autofagia , Neurônios/metabolismo , Lisossomos/metabolismo
17.
Mol Biol Cell ; 35(5): re1, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38598299

RESUMO

Neurofilaments (NFs) are multisubunit, neuron-specific intermediate filaments consisting of a 10-nm diameter filament "core" surrounded by a layer of long intrinsically disordered protein (IDP) "tails." NFs are thought to regulate axonal caliber during development and then stabilize the mature axon, with NF subunit misregulation, mutation, and aggregation featuring prominently in multiple neurological diseases. The field's understanding of NF structure, mechanics, and function has been deeply informed by a rich variety of biochemical, cell biological, and mouse genetic studies spanning more than four decades. These studies have contributed much to our collective understanding of NF function in axonal physiology and disease. In recent years, however, there has been a resurgence of interest in NF subunit proteins in two new contexts: as potential blood- and cerebrospinal fluid-based biomarkers of neuronal damage, and as model IDPs with intriguing properties. Here, we review established principles and more recent discoveries in NF structure and function. Where possible, we place these findings in the context of biophysics of NF assembly, interaction, and contributions to axonal mechanics.


Assuntos
Axônios , Filamentos Intermediários , Proteínas de Neurofilamentos , Filamentos Intermediários/metabolismo , Filamentos Intermediários/fisiologia , Humanos , Animais , Axônios/metabolismo , Axônios/fisiologia , Proteínas de Neurofilamentos/metabolismo , Fenômenos Biomecânicos , Proteínas Intrinsicamente Desordenadas/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Biofísica/métodos , Neurônios/metabolismo , Neurônios/fisiologia
18.
Mol Biol Cell ; 35(6): ar81, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38598291

RESUMO

Neurons are polarized and typically extend multiple dendrites and one axon. To maintain polarity, vesicles carrying dendritic proteins are arrested upon entering the axon. To determine whether kinesin regulation is required for terminating anterograde axonal transport, we overexpressed the dendrite-selective kinesin KIF13A. This caused mistargeting of dendrite-selective vesicles to the axon and a loss of dendritic polarity. Polarity was not disrupted if the kinase MARK2/Par1b was coexpressed. MARK2/Par1b is concentrated in the proximal axon, where it maintains dendritic polarity-likely by phosphorylating S1371 of KIF13A, which lies in a canonical 14-3-3 binding motif. We probed for interactions of KIF13A with 14-3-3 isoforms and found that 14-3-3ß and 14-3-3ζ bound KIF13A. Disruption of MARK2 or 14-3-3 activity by small molecule inhibitors caused a loss of dendritic polarity. These data show that kinesin regulation is integral for dendrite-selective transport. We propose a new model in which KIF13A that moves dendrite-selective vesicles in the proximal axon is phosphorylated by MARK2. Phosphorylated KIF13A is then recognized by 14-3-3, which causes dissociation of KIF13A from the vesicle and termination of transport. These findings define a new paradigm for the regulation of vesicle transport by localized kinesin tail phosphorylation, to restrict dendrite-selective vesicles from entering the axon.


Assuntos
Proteínas 14-3-3 , Axônios , Dendritos , Cinesinas , Cinesinas/metabolismo , Dendritos/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Axônios/metabolismo , Fosforilação , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , Polaridade Celular/fisiologia , Transporte Axonal/fisiologia , Ratos , Neurônios/metabolismo
19.
J Comp Neurol ; 532(3): e25599, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38488687

RESUMO

During embryonic development, the olfactory placode (OP) generates migratory neurons, including olfactory pioneer neurons, cells of the terminal nerve (TN), gonadotropin-releasing hormone-1 (GnRH-1) neurons, and other uncharacterized neurons. Pioneer neurons from the OP induce olfactory bulb (OB) morphogenesis. In mice, GnRH-1 neurons appear in the olfactory system around mid-gestation and migrate via the TN axons to different brain regions. The GnRH-1 neurons are crucial in controlling the hypothalamic-pituitary-gonadal axis. Kallmann syndrome is characterized by impaired olfactory system development, defective OBs, secretion of GnRH-1, and infertility. The precise mechanistic link between the olfactory system and GnRH-1 development remains unclear. Studies in humans and mice highlight the importance of the prokineticin-2/prokineticin-receptor-2 (Prokr2) signaling pathway in OB morphogenesis and GnRH-1 neuronal migration. Prokr2 loss-of-function mutations can cause Kallmann syndrome (KS), and hence the Prokr2 signaling pathway represents a unique model to decipher the olfactory/GnRH-1 connection. We discovered that Prokr2 is expressed in the TN neurons during the critical period of GnRH-1 neuron formation, migration, and induction of OB morphogenesis. Single-cell RNA sequencing identified that the TN is formed by neurons distinct from the olfactory neurons. The TN neurons express multiple genes associated with KS. Our study suggests that the aberrant development of pioneer/TN neurons might cause the KS spectrum.


Assuntos
Síndrome de Kallmann , Humanos , Animais , Camundongos , Síndrome de Kallmann/genética , Síndrome de Kallmann/metabolismo , Neurônios/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Encéfalo/metabolismo , Axônios/metabolismo , Bulbo Olfatório/metabolismo , Movimento Celular/fisiologia
20.
J Cell Sci ; 137(8)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38525600

RESUMO

In neurons, the microtubule (MT) cytoskeleton forms the basis for long-distance protein transport from the cell body into and out of dendrites and axons. To maintain neuronal polarity, the axon initial segment (AIS) serves as a physical barrier, separating the axon from the somatodendritic compartment and acting as a filter for axonal cargo. Selective trafficking is further instructed by axonal enrichment of MT post-translational modifications, which affect MT dynamics and the activity of motor proteins. Here, we compared two knockout mouse lines lacking the respective enzymes for MT tyrosination and detyrosination, and found that both knockouts led to a shortening of the AIS. Neurons from both lines also showed an increased immobile fraction of endolysosomes present in the axon, whereas mobile organelles displayed shortened run distances in the retrograde direction. Overall, our results highlight the importance of maintaining the balance of tyrosinated and detyrosinated MTs for proper AIS length and axonal transport processes.


Assuntos
Transporte Axonal , Lisossomos , Camundongos Knockout , Microtúbulos , Tirosina , Animais , Microtúbulos/metabolismo , Tirosina/metabolismo , Lisossomos/metabolismo , Camundongos , Axônios/metabolismo , Endossomos/metabolismo , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA