Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54.170
Filtrar
1.
Sci Signal ; 17(843): eadr3505, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38954639

RESUMO

Opioids trigger myelin insulation of reward circuit axons in a feedforward loop of addiction.


Assuntos
Analgésicos Opioides , Humanos , Analgésicos Opioides/farmacologia , Animais , Axônios/metabolismo , Axônios/fisiologia , Bainha de Mielina/metabolismo , Recompensa , Transtornos Relacionados ao Uso de Opioides
2.
Elife ; 132024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38949652

RESUMO

Tubulin posttranslational modifications (PTMs) modulate the dynamic properties of microtubules and their interactions with other proteins. However, the effects of tubulin PTMs were often revealed indirectly through the deletion of modifying enzymes or the overexpression of tubulin mutants. In this study, we directly edited the endogenous tubulin loci to install PTM-mimicking or -disabling mutations and studied their effects on microtubule stability, neurite outgrowth, axonal regeneration, cargo transport, and sensory functions in the touch receptor neurons of Caenorhabditis elegans. We found that the status of ß-tubulin S172 phosphorylation and K252 acetylation strongly affected microtubule dynamics, neurite growth, and regeneration, whereas α-tubulin K40 acetylation had little influence. Polyglutamylation and detyrosination in the tubulin C-terminal tail had more subtle effects on microtubule stability likely by modulating the interaction with kinesin-13. Overall, our study systematically assessed and compared several tubulin PTMs for their impacts on neuronal differentiation and regeneration and established an in vivo platform to test the function of tubulin PTMs in neurons.


Assuntos
Caenorhabditis elegans , Microtúbulos , Processamento de Proteína Pós-Traducional , Tubulina (Proteína) , Animais , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Microtúbulos/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Acetilação , Axônios/metabolismo , Axônios/fisiologia , Fosforilação , Regeneração Nervosa , Cinesinas/metabolismo , Cinesinas/genética
3.
Commun Biol ; 7(1): 796, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38951162

RESUMO

The highly complex structure of the brain requires an approach that can unravel its connectivity. Using volume electron microscopy and a dedicated software we can trace and measure all nerve fibers present within different samples of brain tissue. With this software tool, individual dendrites and axons are traced, obtaining a simplified "skeleton" of each fiber, which is linked to its corresponding synaptic contacts. The result is an intricate meshwork of axons and dendrites interconnected by a cloud of synaptic junctions. To test this methodology, we apply it to the stratum radiatum of the hippocampus and layers 1 and 3 of the somatosensory cortex of the mouse. We find that nerve fibers are densely packed in the neuropil, reaching up to 9 kilometers per cubic mm. We obtain the number of synapses, the number and lengths of dendrites and axons, the linear densities of synapses established by dendrites and axons, and their location on dendritic spines and shafts. The quantitative data obtained through this method enable us to identify subtle traits and differences in the synaptic organization of the samples, which might have been overlooked in a qualitative analysis.


Assuntos
Microscopia Eletrônica , Fibras Nervosas , Sinapses , Animais , Camundongos , Microscopia Eletrônica/métodos , Fibras Nervosas/ultraestrutura , Sinapses/ultraestrutura , Axônios/ultraestrutura , Dendritos/ultraestrutura , Encéfalo/ultraestrutura , Córtex Somatossensorial/ultraestrutura , Camundongos Endogâmicos C57BL , Masculino , Software , Hipocampo/ultraestrutura , Hipocampo/citologia , Microscopia Eletrônica de Volume
4.
J Comp Neurol ; 532(7): e25652, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38962882

RESUMO

Although the mammalian cerebral cortex is most often described as a hexalaminar structure, there are cortical areas (primary motor cortex) and species (elephants, cetaceans, and hippopotami), where a cytoarchitecturally indistinct, or absent, layer 4 is noted. Thalamocortical projections from the core, or first order, thalamic system terminate primarily in layers 4/inner 3. We explored the termination sites of core thalamocortical projections in cortical areas and in species where there is no cytoarchitecturally distinct layer 4 using the immunolocalization of vesicular glutamate transporter 2, a known marker of core thalamocortical axon terminals, in 31 mammal species spanning the eutherian radiation. Several variations from the canonical cortical column outline of layer 4 and core thalamocortical inputs were noted. In shrews/microchiropterans, layer 4 was present, but many core thalamocortical projections terminated in layer 1 in addition to layers 4 and inner 3. In primate primary visual cortex, the sublaminated layer 4 was associated with a specialized core thalamocortical projection pattern. In primate primary motor cortex, no cytoarchitecturally distinct layer 4 was evident and the core thalamocortical projections terminated throughout layer 3. In the African elephant, cetaceans, and river hippopotamus, no cytoarchitecturally distinct layer 4 was observed and core thalamocortical projections terminated primarily in inner layer 3 and less densely in outer layer 3. These findings are contextualized in terms of cortical processing, perception, and the evolutionary trajectory leading to an indistinct or absent cortical layer 4.


Assuntos
Axônios , Neocórtex , Vias Neurais , Tálamo , Animais , Tálamo/citologia , Tálamo/anatomia & histologia , Neocórtex/citologia , Neocórtex/anatomia & histologia , Vias Neurais/citologia , Vias Neurais/anatomia & histologia , Axônios/fisiologia , Mamíferos/anatomia & histologia , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Especificidade da Espécie
5.
Sci Adv ; 10(27): eado9120, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38959311

RESUMO

A bioinspired hydrogel composed of hyaluronic acid-graft-dopamine (HADA) and a designer peptide HGF-(RADA)4-DGDRGDS (HRR) was presented to enhance tissue integration following spinal cord injury (SCI). The HADA/HRR hydrogel manipulated the infiltration of PDGFRß+ cells in a parallel pattern, transforming dense scars into an aligned fibrous substrate that guided axonal regrowth. Further incorporation of NT3 and curcumin promoted axonal regrowth and survival of interneurons at lesion borders, which served as relays for establishing heterogeneous axon connections in a target-specific manner. Notable improvements in motor, sensory, and bladder functions resulted in rats with complete spinal cord transection. The HADA/HRR + NT3/Cur hydrogel promoted V2a neuron accumulation in ventral spinal cord, facilitating the recovery of locomotor function. Meanwhile, the establishment of heterogeneous neural connections across the hemisected lesion of canines was documented in a target-specific manner via neuronal relays, significantly improving motor functions. Therefore, biomaterials can inspire beneficial biological activities for SCI repair.


Assuntos
Matriz Extracelular , Hidrogéis , Traumatismos da Medula Espinal , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Animais , Hidrogéis/química , Ratos , Matriz Extracelular/metabolismo , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Cães , Axônios/metabolismo , Axônios/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Ácido Hialurônico/química , Ácido Hialurônico/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Dopamina/metabolismo , Feminino , Modelos Animais de Doenças , Ratos Sprague-Dawley , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Medula Espinal/metabolismo
6.
J Biomed Sci ; 31(1): 69, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992696

RESUMO

BACKGROUND: Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity. METHODS: Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles. RESULTS: Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles. CONCLUSIONS: We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.


Assuntos
Plasticidade Neuronal , Proteínas de Ligação a RNA , Transmissão Sináptica , Proteína Vesicular 2 de Transporte de Glutamato , Animais , Camundongos , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Plasticidade Neuronal/fisiologia , Transmissão Sináptica/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Camundongos Knockout , Axônios/metabolismo , Axônios/fisiologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Masculino , Biossíntese de Proteínas
7.
Cells ; 13(13)2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38995011

RESUMO

Unsuccessful axonal regeneration in transected spinal cord injury (SCI) is mainly attributed to shortage of growth factors, inhibitory glial scar, and low intrinsic regenerating capacity of severely injured neurons. Previously, we constructed an axonal growth permissive pathway in a thoracic hemisected injury by transplantation of Schwann cells overexpressing glial-cell-derived neurotrophic factor (SCs-GDNF) into the lesion gap as well as the caudal cord and proved that this novel permissive bridge promoted the regeneration of descending propriospinal tract (dPST) axons across and beyond the lesion. In the current study, we subjected rats to complete thoracic (T11) spinal cord transections and examined whether these combinatorial treatments can support dPST axons' regeneration beyond the transected injury. The results indicated that GDNF significantly improved graft-host interface by promoting integration between SCs and astrocytes, especially the migration of reactive astrocyte into SCs-GDNF territory. The glial response in the caudal graft area has been significantly attenuated. The astrocytes inside the grafted area were morphologically characterized by elongated and slim process and bipolar orientation accompanied by dramatically reduced expression of glial fibrillary acidic protein. Tremendous dPST axons have been found to regenerate across the lesion and back to the caudal spinal cord which were otherwise difficult to see in control groups. The caudal synaptic connections were formed, and regenerated axons were remyelinated. The hindlimb locomotor function has been improved.


Assuntos
Axônios , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Regeneração Nervosa , Células de Schwann , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Células de Schwann/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Axônios/metabolismo , Ratos , Ratos Sprague-Dawley , Feminino , Astrócitos/metabolismo
8.
Alzheimers Res Ther ; 16(1): 146, 2024 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961441

RESUMO

BACKGROUND: Increasing evidence supports the use of plasma biomarkers of amyloid, tau, neurodegeneration, and neuroinflammation for diagnosis of dementia. However, their performance for positive and differential diagnosis of dementia with Lewy bodies (DLB) in clinical settings is still uncertain. METHODS: We conducted a retrospective biomarker study in two tertiary memory centers, Paris Lariboisière and CM2RR Strasbourg, France, enrolling patients with DLB (n = 104), Alzheimer's disease (AD, n = 76), and neurological controls (NC, n = 27). Measured biomarkers included plasma Aß40/Aß42 ratio, p-tau181, NfL, and GFAP using SIMOA and plasma YKL-40 and sTREM2 using ELISA. DLB patients with available CSF analysis (n = 90) were stratified according to their CSF Aß profile. RESULTS: DLB patients displayed modified plasma Aß ratio, p-tau181, and GFAP levels compared with NC and modified plasma Aß ratio, p-tau181, GFAP, NfL, and sTREM2 levels compared with AD patients. Plasma p-tau181 best differentiated DLB from AD patients (ROC analysis, area under the curve [AUC] = 0.80) and NC (AUC = 0.78), and combining biomarkers did not improve diagnosis performance. Plasma p-tau181 was the best standalone biomarker to differentiate amyloid-positive from amyloid-negative DLB cases (AUC = 0.75) and was associated with cognitive status in the DLB group. Combining plasma Aß ratio, p-tau181 and NfL increased performance to identify amyloid copathology (AUC = 0.79). Principal component analysis identified different segregation patterns of biomarkers in the DLB and AD groups. CONCLUSIONS: Amyloid, tau, neurodegeneration and neuroinflammation plasma biomarkers are modified in DLB, albeit with moderate diagnosis performance. Plasma p-tau181 can contribute to identify Aß copathology in DLB.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Biomarcadores , Doença por Corpos de Lewy , Proteínas tau , Humanos , Doença por Corpos de Lewy/sangue , Doença por Corpos de Lewy/líquido cefalorraquidiano , Doença por Corpos de Lewy/diagnóstico , Proteínas tau/sangue , Proteínas tau/líquido cefalorraquidiano , Feminino , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Masculino , Idoso , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Estudos Retrospectivos , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/diagnóstico , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Axônios/patologia , Doenças Neuroinflamatórias/sangue , Doenças Neuroinflamatórias/diagnóstico , Doenças Neuroinflamatórias/líquido cefalorraquidiano , Proteína 1 Semelhante à Quitinase-3/sangue , Proteína 1 Semelhante à Quitinase-3/líquido cefalorraquidiano , Proteína Glial Fibrilar Ácida/sangue , Proteína Glial Fibrilar Ácida/líquido cefalorraquidiano , Proteínas de Neurofilamentos/sangue , Proteínas de Neurofilamentos/líquido cefalorraquidiano , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/líquido cefalorraquidiano , Receptores Imunológicos/sangue , Diagnóstico Diferencial , Glicoproteínas de Membrana
9.
Int J Mol Sci ; 25(13)2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-39000434

RESUMO

GRT-X, which targets both the mitochondrial translocator protein (TSPO) and the Kv7.2/3 (KCNQ2/3) potassium channels, has been shown to efficiently promote recovery from cervical spine injury. In the present work, we investigate the role of GRT-X and its two targets in the axonal growth of dorsal root ganglion (DRG) neurons. Neurite outgrowth was quantified in DRG explant cultures prepared from wild-type C57BL6/J and TSPO-KO mice. TSPO was pharmacologically targeted with the agonist XBD173 and the Kv7 channels with the activator ICA-27243 and the inhibitor XE991. GRT-X efficiently stimulated DRG axonal growth at 4 and 8 days after its single administration. XBD173 also promoted axonal elongation, but only after 8 days and its repeated administration. In contrast, both ICA27243 and XE991 tended to decrease axonal elongation. In dissociated DRG neuron/Schwann cell co-cultures, GRT-X upregulated the expression of genes associated with axonal growth and myelination. In the TSPO-KO DRG cultures, the stimulatory effect of GRT-X on axonal growth was completely lost. However, GRT-X and XBD173 activated neuronal and Schwann cell gene expression after TSPO knockout, indicating the presence of additional targets warranting further investigation. These findings uncover a key role of the dual mode of action of GRT-X in the axonal elongation of DRG neurons.


Assuntos
Axônios , Gânglios Espinais , Receptores de GABA , Animais , Gânglios Espinais/metabolismo , Gânglios Espinais/citologia , Camundongos , Axônios/metabolismo , Receptores de GABA/metabolismo , Receptores de GABA/genética , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ2/genética , Camundongos Knockout , Camundongos Endogâmicos C57BL , Células Cultivadas , Células de Schwann/metabolismo , Células de Schwann/efeitos dos fármacos , Células de Schwann/citologia , Técnicas de Cocultura , Neurônios/metabolismo , Neurônios/efeitos dos fármacos
10.
Int J Mol Sci ; 25(13)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39000515

RESUMO

Advanced glycation end-products (AGEs) form through non-enzymatic glycation of various proteins. Optic nerve degeneration is a frequent complication of diabetes, and retinal AGE accumulation is strongly linked to the development of diabetic retinopathy. Type 2 diabetes mellitus is a major risk factor for Alzheimer's disease (AD), with patients often exhibiting optic axon degeneration in the nerve fiber layer. Notably, a gap exists in our understanding of how AGEs contribute to neuronal degeneration in the optic nerve within the context of both diabetes and AD. Our previous work demonstrated that glyceraldehyde (GA)-derived toxic advanced glycation end-products (TAGE) disrupt neurite outgrowth through TAGE-ß-tubulin aggregation and tau phosphorylation in neural cultures. In this study, we further illustrated GA-induced suppression of optic nerve axonal elongation via abnormal ß-tubulin aggregation in mouse retinas. Elucidating this optic nerve degeneration mechanism holds promise for bridging the knowledge gap regarding vision loss associated with diabetes mellitus and AD.


Assuntos
Axônios , Produtos Finais de Glicação Avançada , Nervo Óptico , Tubulina (Proteína) , Animais , Tubulina (Proteína)/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Camundongos , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Nervo Óptico/efeitos dos fármacos , Axônios/metabolismo , Axônios/efeitos dos fármacos , Axônios/patologia , Camundongos Endogâmicos C57BL , Agregados Proteicos/efeitos dos fármacos
11.
Proc Natl Acad Sci U S A ; 121(31): e2402755121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39042673

RESUMO

The precise assembly of a functional nervous system relies on axon guidance cues. Beyond engaging their cognate receptors and initiating signaling cascades that modulate cytoskeletal dynamics, guidance cues also bind components of the extracellular matrix, notably proteoglycans, yet the role and mechanisms of these interactions remain poorly understood. We found that Drosophila secreted semaphorins bind specifically to glycosaminoglycan (GAG) chains of proteoglycans, showing a preference based on the degree of sulfation. Structural analysis of Sema2b unveiled multiple GAG-binding sites positioned outside canonical plexin-binding site, with the highest affinity binding site located at the C-terminal tail, characterized by a lysine-rich helical arrangement that appears to be conserved across secreted semaphorins. In vivo studies revealed a crucial role of the Sema2b C-terminal tail in specifying the trajectory of olfactory receptor neurons. We propose that secreted semaphorins tether to the cell surface through interactions with GAG chains of proteoglycans, facilitating their presentation to cognate receptors on passing axons.


Assuntos
Orientação de Axônios , Proteínas de Drosophila , Proteoglicanas , Semaforinas , Transdução de Sinais , Animais , Semaforinas/metabolismo , Semaforinas/genética , Proteoglicanas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Axônios/metabolismo , Drosophila melanogaster/metabolismo , Glicosaminoglicanos/metabolismo , Sítios de Ligação , Ligação Proteica , Neurônios Receptores Olfatórios/metabolismo
12.
Int J Mol Sci ; 25(13)2024 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-39000003

RESUMO

Peripheral nerve injuries (PNIs) represent a significant clinical challenge, particularly in elderly populations where axonal remyelination and regeneration are impaired. Developing therapies to enhance these processes is crucial for improving PNI repair outcomes. Glutamate carboxypeptidase II (GCPII) is a neuropeptidase that plays a pivotal role in modulating glutamate signaling through its enzymatic cleavage of the abundant neuropeptide N-acetyl aspartyl glutamate (NAAG) to liberate glutamate. Within the PNS, GCPII is expressed in Schwann cells and activated macrophages, and its expression is amplified with aging. In this study, we explored the therapeutic potential of inhibiting GCPII activity following PNI. We report significant GCPII protein and activity upregulation following PNI, which was normalized by the potent and selective GCPII inhibitor 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). In vitro, 2-PMPA robustly enhanced myelination in dorsal root ganglion (DRG) explants. In vivo, using a sciatic nerve crush injury model in aged mice, 2-PMPA accelerated remyelination, as evidenced by increased myelin sheath thickness and higher numbers of remyelinated axons. These findings suggest that GCPII inhibition may be a promising therapeutic strategy to enhance remyelination and potentially improve functional recovery after PNI, which is especially relevant in elderly PNI patients where this process is compromised.


Assuntos
Glutamato Carboxipeptidase II , Traumatismos dos Nervos Periféricos , Remielinização , Animais , Camundongos , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Traumatismos dos Nervos Periféricos/metabolismo , Remielinização/efeitos dos fármacos , Glutamato Carboxipeptidase II/antagonistas & inibidores , Glutamato Carboxipeptidase II/metabolismo , Bainha de Mielina/metabolismo , Bainha de Mielina/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Camundongos Endogâmicos C57BL , Regeneração Nervosa/efeitos dos fármacos , Nervo Isquiático/lesões , Nervo Isquiático/efeitos dos fármacos , Masculino , Axônios/efeitos dos fármacos , Axônios/metabolismo
13.
J Nanobiotechnology ; 22(1): 399, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38970101

RESUMO

Spinal cord injury (SCI) represents a profound central nervous system affliction, resulting in irreversibly compromised daily activities and disabilities. SCI involves excessive inflammatory responses, which are characterized by the existence of high levels of proinflammatory M1 macrophages, and neuronal mitochondrial energy deficit, exacerbating secondary damage and impeding axon regeneration. This study delves into the mechanistic intricacies of SCI, offering insights from the perspectives of neuroimmune regulation and mitochondrial function, leading to a pro-fibrotic macrophage phenotype and energy-supplying deficit. To address these challenges, we developed a smart scaffold incorporating enzyme mimicry nanoparticle-ceriumoxide (COPs) into nanofibers (NS@COP), which aims to pioneer a targeted neuroimmune repair strategy, rescuing CGRP receptor on macrophage and concurrently remodeling mitochondrial function. Our findings indicate that the integrated COPs restore the responsiveness of pro-inflammatory macrophages to calcitonin gene-related peptide (CGRP) signal by up-regulating receptor activity modifying protein 1 (RAMP1), a vital component of the CGRP receptor. This promotes macrophage fate commitment to an anti-inflammatory pro-resolution M2 phenotype, then alleviating glial scar formation. In addition, NS@COP implantation also protected neuronal mitochondrial function. Collectively, our results suggest that the strategy of integrating nanozyme COP nanoparticles into a nanofiber scaffold provides a promising therapeutic candidate for spinal cord trauma via rational regulation of neuroimmune communication and mitochondrial function.


Assuntos
Axônios , Macrófagos , Nanofibras , Regeneração Nervosa , Traumatismos da Medula Espinal , Animais , Axônios/metabolismo , Nanofibras/química , Regeneração Nervosa/efeitos dos fármacos , Camundongos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Ratos , Alicerces Teciduais/química , Nanopartículas/química , Ratos Sprague-Dawley , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Feminino , Camundongos Endogâmicos C57BL
14.
J Neurogenet ; 38(2): 35-40, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38975976

RESUMO

Pathogenic, biallelic variants in SORD were identified in 2020 as a novel cause for autosomal-recessive Charcot-Marie-Tooth disease (CMT) type 2, an inherited neuropathy. SORD codes for the enzyme sorbitol dehydrogenase. Loss of this enzyme's activity leads to an increase of sorbitol in serum. We retrospectively screened 166 patients with axonal neuropathy (predominantly CMT type 2, but including intermediate form of CMT and distal hereditary motor neuropathy (dHMN)) without identified genetic etiology for SORD mutations at a single large German neuromuscular center. Clinical and electrophysiology exam findings were analyzed for genotype-phenotype correlation. Five patients of the total cohort of 166 patients harbored pathogenic variants in SORD (3%). The homozygous frameshift variant c.757delG (p.Ala253Glnfs*27) was the most common (4/5). One additional case carried this variant on one allele only and an additional pathogenic missense variant c.458C > A (p.Ala153Asp) on the other allele. Age of onset ranged from early infancy to mid-twenties, and phenotypes comprised axonal CMT (4) and dHMN (1). Our findings strengthen the importance of screening for pathogenic variants in SORD, especially in patients with genetically unconfirmed axonal neuropathy, especially CMT type 2 and dHMN.


Assuntos
Doença de Charcot-Marie-Tooth , Fenótipo , Humanos , Doença de Charcot-Marie-Tooth/genética , Feminino , Masculino , Adulto , Mutação , Estudos Retrospectivos , Estudos de Associação Genética/métodos , Criança , Adolescente , Axônios/patologia , Adulto Jovem , Pré-Escolar
15.
Elife ; 132024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39012795

RESUMO

Axo-axonic cells (AACs), also called chandelier cells (ChCs) in the cerebral cortex, are the most distinctive type of GABAergic interneurons described in the neocortex, hippocampus, and basolateral amygdala (BLA). AACs selectively innervate glutamatergic projection neurons (PNs) at their axon initial segment (AIS), thus may exert decisive control over PN spiking and regulate PN functional ensembles. However, the brain-wide distribution, synaptic connectivity, and circuit function of AACs remain poorly understood, largely due to the lack of specific and reliable experimental tools. Here, we have established an intersectional genetic strategy that achieves specific and comprehensive targeting of AACs throughout the mouse brain based on their lineage (Nkx2.1) and molecular (Unc5b, Pthlh) markers. We discovered that AACs are deployed across essentially all the pallium-derived brain structures, including not only the dorsal pallium-derived neocortex and medial pallium-derived hippocampal formation, but also the lateral pallium-derived claustrum-insular complex, and the ventral pallium-derived extended amygdaloid complex and olfactory centers. AACs are also abundant in anterior olfactory nucleus, taenia tecta, and lateral septum. AACs show characteristic variations in density across neocortical areas and layers and across subregions of the hippocampal formation. Neocortical AACs comprise multiple laminar subtypes with distinct dendritic and axonal arborization patterns. Retrograde monosynaptic tracing from AACs across neocortical, hippocampal, and BLA regions reveal shared as well as distinct patterns of synaptic input. Specific and comprehensive targeting of AACs facilitates the study of their developmental genetic program and circuit function across brain structures, providing a ground truth platform for understanding the conservation and variation of a bona fide cell type across brain regions and species.


Whether we are memorising facts or reacting to a loud noise, nerve cells in different brain areas must be able to communicate with one another through precise, meaningful signals. Specialized nerve cells known as interneurons act as "traffic lights" to precisely regulate when and where this information flows in neural circuits. Axo-axonic cells are a rare type of inhibitory interneuron that are thought to be particularly important for controlling the passage of information between different groups of excitatory neurons. This is because they only connect to one key part of their target cell ­ the axon-initial segment ­ where the electrical signals needed for brain communication (known as action potentials) are initiated. Since axo-axonic cells are inhibitory interneurons, this connection effectively allows them to 'veto' the generation of these signals at their source. Although axo-axonic cells have been identified in three brain regions using traditional anatomical methods, there were no 'tags' readily available that can reliably identify them. Therefore, much about these cells remained unknown, including how widespread they are in the mammalian brain. To solve this problem, Raudales et al. investigated which genes are switched on in axo-axonic cells but not in other cells, identifying a unique molecular signature that could be used to mark, record, and manipulate these cells. Microscopy imaging of brain tissue from mice in which axo-axonic cells had been identified revealed that they are present in many more brain areas than previously thought, including nearly all regions of the broadly defined cerebral cortex and even the hypothalamus, which controls many innate behaviors. Axo-axonic cells were also 'wired up' differently, depending on where they were located; for example, those in brain areas associated with memory and emotions had wider-ranging input connections than other areas. The finding of Raudales et al. provide, for the first time, a method to directly track and manipulate axo-axonic cells in the brain. Since dysfunction in axo-axonic cells is also associated with neurological disorders like epilepsy and schizophrenia, gaining an insight into their distribution and connectivity could help to develop better treatments for these conditions.


Assuntos
Neurônios GABAérgicos , Interneurônios , Animais , Interneurônios/fisiologia , Interneurônios/metabolismo , Neurônios GABAérgicos/fisiologia , Neurônios GABAérgicos/metabolismo , Camundongos , Encéfalo/fisiologia , Encéfalo/citologia , Sinapses/fisiologia , Sinapses/metabolismo , Axônios/fisiologia , Axônios/metabolismo , Masculino
16.
Mol Biol Cell ; 35(8): ar109, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38985523

RESUMO

The Drosophila RNA-binding protein (RBP) Nab2 acts in neurons to regulate neurodevelopment and is orthologous to the human intellectual disability-linked RBP, ZC3H14. Nab2 governs axon projection in mushroom body neurons and limits dendritic arborization of class IV sensory neurons in part by regulating splicing events in ∼150 mRNAs. Analysis of the Sex-lethal (Sxl) mRNA revealed that Nab2 promotes an exon-skipping event and regulates m6A methylation on Sxl pre-mRNA by the Mettl3 methyltransferase. Mettl3 heterozygosity broadly rescues Nab2null phenotypes implying that Nab2 acts through similar mechanisms on other RNAs, including unidentified targets involved in neurodevelopment. Here, we show that Nab2 and Mettl3 regulate the removal of a 5'UTR (untranslated region) intron in the trio pre-mRNA. Trio utilizes two GEF domains to balance Rac and RhoGTPase activity. Intriguingly, an isoform of Trio containing only the RhoGEF domain, GEF2, is depleted in Nab2null nervous tissue. Expression of Trio-GEF2 rescues projection defects in Nab2null axons and dendrites, while the GEF1 Rac1-regulatory domain exacerbates these defects, suggesting Nab2-mediated regulation Trio-GEF activities. Collectively, these data indicate that Nab2-regulated processing of trio is critical for balancing Trio-GEF1 and -GEF2 activity and show that Nab2, Mettl3, and Trio function in a common pathway that shapes axon and dendrite morphology.


Assuntos
Axônios , Dendritos , Proteínas de Drosophila , Drosophila melanogaster , Fatores de Troca do Nucleotídeo Guanina , Proteínas de Ligação a RNA , Animais , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Axônios/metabolismo , Dendritos/metabolismo , Drosophila melanogaster/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Metiltransferases/metabolismo , Metiltransferases/genética , Splicing de RNA , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Precursores de RNA/metabolismo , Precursores de RNA/genética
17.
Zhen Ci Yan Jiu ; 49(7): 767-776, 2024 Jul 25.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-39020496

RESUMO

Stroke brings the pathological changes of brain tissues such as hematoma formation or ischemia and hypoxia, which leads to neuronal death and axon degeneration. Axon regeneration after its injury is mainly dependent on the surrounding microenvironment and the related proteins, including glial scar, myelin associated inhibitory factors, axon guidance molecules, and neurotrophic factors. All of them affect axon growth by regulating the morphology and orientation of growth cones, the synaptic stability, and the proliferation and differentiation of neural stem cells. This article summarizes the mechanism of acupuncture in regulating axon regeneration after stroke. Acupuncture inhibits glial scar formation, alleviates the inhibitory effects of its physical and chemical barriers on axon growth, reverses the inhibitory effects of myelin-related inhibitory factors on axon growth, and adjusts the level of axon guidance molecules to promote the proliferation and differentiation of neural stem cells and the regeneration of injured axons, and up-regulates neurotrophic factors. Eventually, post-stroke nerve injury can be ameliorated.


Assuntos
Terapia por Acupuntura , Axônios , Regeneração Nervosa , Acidente Vascular Cerebral , Humanos , Animais , Axônios/metabolismo , Axônios/fisiologia , Acidente Vascular Cerebral/terapia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Células-Tronco Neurais/metabolismo
18.
Nat Commun ; 15(1): 6068, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39025931

RESUMO

Neurexins are key adhesion proteins that coordinate extracellular and intracellular synaptic components. Nonetheless, the low abundance of these multidomain proteins has complicated any localization and structure-function studies. Here we combine an ALFA tag (AT)/nanobody (NbALFA) tool with classic genetics, cell biology and electrophysiology to examine the distribution and function of the Drosophila Nrx-1 in vivo. We generate full-length and ΔPDZ ALFA-tagged Nrx-1 variants and find that the PDZ binding motif is key to Nrx-1 surface expression. A PDZ binding motif provided in trans, via genetically encoded cytosolic NbALFA-PDZ chimera, fully restores the synaptic localization and function of NrxΔPDZ-AT. Using cytosolic NbALFA-mScarlet intrabody, we achieve compartment-specific detection of endogenous Nrx-1, track live Nrx-1 transport along the motor neuron axons, and demonstrate that Nrx-1 co-migrates with Rab2-positive vesicles. Our findings illustrate the versatility of the ALFA system and pave the way towards dissecting functional domains of complex proteins in vivo.


Assuntos
Proteínas de Drosophila , Anticorpos de Domínio Único , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Anticorpos de Domínio Único/metabolismo , Drosophila melanogaster/metabolismo , Neurônios Motores/metabolismo , Domínios PDZ , Axônios/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/genética , Transporte Proteico , Moléculas de Adesão Celular Neuronais
19.
Sci Rep ; 14(1): 16643, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39025960

RESUMO

Circuit implementations of neuronal networks so far have been focusing on synaptic weight changes as network growth principles. Besides these weight changes, however, it is also useful to incorporate additional network growth principles such as guided axon growth and pruning. These allow for dynamical signal delays and a higher degree of self-organization, and can thus lead to novel circuit design principles. In this work we develop an ideal, bio-inspired electrical circuit mimicking growth and pruning controlled by guidance cues. The circuit is based on memristively coupled neuronal oscillators. As coupling element, we use memsensors consisting of a general sensor, two gradient sensors, and two memristors. The oscillators and memsensors are arranged in a grid structure, where oscillators and memsensors realize nodes and edges, respectively. This allows for arbitrary 2D growth scenarios with axon growth controlled by guidance cues. Simulation results show that the circuit successfully mimics a biological example in which two neurons initially grow towards two target neurons, where undesired connections are pruned later on.


Assuntos
Axônios , Modelos Neurológicos , Rede Nervosa , Axônios/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Simulação por Computador , Animais , Humanos
20.
Exp Eye Res ; 245: 109988, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38964496

RESUMO

Autism spectrum disorder (ASD) is a group of neurodevelopment disorders characterized by deficits in social interaction and communication, and repetitive or stereotyped behavior. Autistic children are more likely to have vision problems, and ASD is unusually common among blind people. However, the mechanisms behind the vision disorders in autism are unclear. Stabilizing WNT-targeted scaffold protein Axin2 by XAV939 during embryonic development causes overproduction of cortical neurons and leads to autistic-like behaviors in mice. In this study, we investigated the relationship between vision abnormality and autism using an XAV939-induced mouse model of autism. We found that the mice receiving XAV939 had decreased amplitude of bright light-adaptive ERG. The amplitudes and latency of flash visual evoked potential recorded from XAV939-treated mice were lower and longer, respectively than in the control mice, suggesting that XAV939 inhibits visual signal processing and conductance. Anatomically, the diameters of RGC axons were reduced when Axin2 was stabilized during the development, and the optic fibers had defective myelin sheaths and reduced oligodendrocytes. The results suggest that the WNT signaling pathway is crucial for optic nerve development. This study provides experimental evidence that conditions interfering with brain development may also lead to visual problems, which in turn might exaggerate the autistic features in humans.


Assuntos
Proteína Axina , Modelos Animais de Doenças , Potenciais Evocados Visuais , Nervo Óptico , Animais , Proteína Axina/metabolismo , Camundongos , Potenciais Evocados Visuais/fisiologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Eletrorretinografia , Camundongos Endogâmicos C57BL , Axônios/patologia , Células Ganglionares da Retina/patologia , Células Ganglionares da Retina/metabolismo , Masculino , Via de Sinalização Wnt/fisiologia , Transtorno do Espectro Autista/fisiopatologia , Transtorno do Espectro Autista/metabolismo , Transtorno Autístico/fisiopatologia , Transtorno Autístico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA