Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 292.702
Filtrar
1.
J Neuroinflammation ; 21(1): 119, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38715061

RESUMO

BACKGROUND: Cerebral malaria (CM) is the most lethal complication of malaria, and survivors usually endure neurological sequelae. Notably, the cytotoxic effect of infiltrating Plasmodium-activated CD8+ T cells on cerebral microvasculature endothelial cells is a prominent feature of the experimental CM (ECM) model with blood-brain barrier disruption. However, the damage effect of CD8+ T cells infiltrating the brain parenchyma on neurons remains unclear. Based on the immunosuppressive effect of the PD-1/PD-L1 pathway on T cells, our previous study demonstrated that the systemic upregulation of PD-L1 to inhibit CD8+ T cell function could effectively alleviate the symptoms of ECM mice. However, it has not been reported whether neurons can suppress the pathogenic effect of CD8+ T cells through the PD-1/PD-L1 negative immunomodulatory pathway. As the important inflammatory factor of CM, interferons can induce the expression of PD-L1 via different molecular mechanisms according to the neuro-immune microenvironment. Therefore, this study aimed to investigate the direct interaction between CD8+ T cells and neurons, as well as the mechanism of neurons to alleviate the pathogenic effect of CD8+ T cells through up-regulating PD-L1 induced by IFNs. METHODS: Using the ECM model of C57BL/6J mice infected with Plasmodium berghei ANKA (PbA), morphological observations were conducted in vivo by electron microscope and IF staining. The interaction between the ECM CD8+ T cells (immune magnetic bead sorting from spleen of ECM mice) and primary cultured cortical neurons in vitro was observed by IF staining and time-lapse photography. RNA-seq was performed to analyze the signaling pathway of PD-L1 upregulation in neurons induced by IFNß or IFNγ, and verified through q-PCR, WB, IF staining, and flow cytometry both in vitro and in vivo using IFNAR or IFNGR gene knockout mice. The protective effect of adenovirus-mediated PD-L1 IgGFc fusion protein expression was verified in ECM mice with brain stereotaxic injection in vivo and in primary cultured neurons via viral infection in vitro. RESULTS: In vivo, ECM mice showed infiltration of activated CD8+ T cells and neuronal injury in the brain parenchyma. In vitro, ECM CD8+ T cells were in direct contact with neurons and induced axonal damage, as an active behavior. The PD-L1 protein level was elevated in neurons of ECM mice and in primary cultured neurons induced by IFNß, IFNγ, or ECM CD8+ T cells in vitro. Furthermore, the IFNß or IFNγ induced neuronal expression of PD-L1 was mediated by increasing STAT1/IRF1 pathway via IFN receptors. The increase of PD-L1 expression in neurons during PbA infection was weakened after deleting the IFNAR or IFNGR. Increased PD-L1 expression by adenovirus partially protected neurons from CD8+ T cell-mediated damage both in vitro and in vivo. CONCLUSION: Our study demonstrates that both type I and type II IFNs can induce neurons to upregulate PD-L1 via the STAT1/IRF1 pathway mediated by IFN receptors to protect against activated CD8+ T cell-mediated damage, providing a targeted pathway to alleviate neuroinflammation during ECM.


Assuntos
Antígeno B7-H1 , Linfócitos T CD8-Positivos , Malária Cerebral , Camundongos Endogâmicos C57BL , Neurônios , Fator de Transcrição STAT1 , Regulação para Cima , Animais , Camundongos , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/metabolismo , Malária Cerebral/imunologia , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Camundongos Knockout , Neurônios/metabolismo , Plasmodium berghei , Transdução de Sinais/fisiologia , Fator de Transcrição STAT1/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
Science ; 384(6696): eadk4858, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38723085

RESUMO

To fully understand how the human brain works, knowledge of its structure at high resolution is needed. Presented here is a computationally intensive reconstruction of the ultrastructure of a cubic millimeter of human temporal cortex that was surgically removed to gain access to an underlying epileptic focus. It contains about 57,000 cells, about 230 millimeters of blood vessels, and about 150 million synapses and comprises 1.4 petabytes. Our analysis showed that glia outnumber neurons 2:1, oligodendrocytes were the most common cell, deep layer excitatory neurons could be classified on the basis of dendritic orientation, and among thousands of weak connections to each neuron, there exist rare powerful axonal inputs of up to 50 synapses. Further studies using this resource may bring valuable insights into the mysteries of the human brain.


Assuntos
Neurônios , Sinapses , Lobo Temporal , Humanos , Neurônios/ultraestrutura , Sinapses/fisiologia , Sinapses/ultraestrutura , Oligodendroglia/citologia , Neuroglia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/citologia , Córtex Cerebral/ultraestrutura , Dendritos/fisiologia , Axônios/fisiologia , Axônios/ultraestrutura
3.
Open Biol ; 14(5): 240018, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38745463

RESUMO

The neuronal cell adhesion molecule contactin-4 (CNTN4) is genetically associated with autism spectrum disorder (ASD) and other psychiatric disorders. Cntn4-deficient mouse models have previously shown that CNTN4 plays important roles in axon guidance and synaptic plasticity in the hippocampus. However, the pathogenesis and functional role of CNTN4 in the cortex has not yet been investigated. Our study found a reduction in cortical thickness in the motor cortex of Cntn4 -/- mice, but cortical cell migration and differentiation were unaffected. Significant morphological changes were observed in neurons in the M1 region of the motor cortex, indicating that CNTN4 is also involved in the morphology and spine density of neurons in the motor cortex. Furthermore, mass spectrometry analysis identified an interaction partner for CNTN4, confirming an interaction between CNTN4 and amyloid-precursor protein (APP). Knockout human cells for CNTN4 and/or APP revealed a relationship between CNTN4 and APP. This study demonstrates that CNTN4 contributes to cortical development and that binding and interplay with APP controls neural elongation. This is an important finding for understanding the physiological function of APP, a key protein for Alzheimer's disease. The binding between CNTN4 and APP, which is involved in neurodevelopment, is essential for healthy nerve outgrowth.


Assuntos
Precursor de Proteína beta-Amiloide , Contactinas , Camundongos Knockout , Neurônios , Animais , Camundongos , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Humanos , Contactinas/metabolismo , Contactinas/genética , Neurônios/metabolismo , Córtex Motor/metabolismo , Ligação Proteica , Movimento Celular
4.
Nat Commun ; 15(1): 3969, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730242

RESUMO

Encephalitis is a rare and potentially fatal manifestation of herpes simplex type 1 infection. Following genome-wide genetic analyses, we identified a previously uncharacterized and very rare heterozygous variant in the E3 ubiquitin ligase WWP2, in a 14-month-old girl with herpes simplex encephalitis. The p.R841H variant (NM_007014.4:c.2522G > A) impaired TLR3 mediated signaling in inducible pluripotent stem cells-derived neural precursor cells and neurons; cells bearing this mutation were also more susceptible to HSV-1 infection compared to control cells. The p.R841H variant increased TRIF ubiquitination in vitro. Antiviral immunity was rescued following the correction of p.R841H by CRISPR-Cas9 technology. Moreover, the introduction of p.R841H in wild type cells reduced such immunity, suggesting that this mutation is linked to the observed phenotypes.


Assuntos
Encefalite por Herpes Simples , Herpesvirus Humano 1 , Mutação , Ubiquitina-Proteína Ligases , Humanos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Feminino , Encefalite por Herpes Simples/genética , Lactente , Herpesvirus Humano 1/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Ubiquitinação , Neurônios/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Sistemas CRISPR-Cas
5.
Nat Commun ; 15(1): 3542, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719802

RESUMO

Understanding the functional connectivity between brain regions and its emergent dynamics is a central challenge. Here we present a theory-experiment hybrid approach involving iteration between a minimal computational model and in vivo electrophysiological measurements. Our model not only predicted spontaneous persistent activity (SPA) during Up-Down-State oscillations, but also inactivity (SPI), which has never been reported. These were confirmed in vivo in the membrane potential of neurons, especially from layer 3 of the medial and lateral entorhinal cortices. The data was then used to constrain two free parameters, yielding a unique, experimentally determined model for each neuron. Analytic and computational analysis of the model generated a dozen quantitative predictions about network dynamics, which were all confirmed in vivo to high accuracy. Our technique predicted functional connectivity; e. g. the recurrent excitation is stronger in the medial than lateral entorhinal cortex. This too was confirmed with connectomics data. This technique uncovers how differential cortico-entorhinal dialogue generates SPA and SPI, which could form an energetically efficient working-memory substrate and influence the consolidation of memories during sleep. More broadly, our procedure can reveal the functional connectivity of large networks and a theory of their emergent dynamics.


Assuntos
Córtex Entorrinal , Modelos Neurológicos , Neurônios , Córtex Entorrinal/fisiologia , Animais , Neurônios/fisiologia , Masculino , Conectoma , Rede Nervosa/fisiologia , Potenciais da Membrana/fisiologia , Vias Neurais/fisiologia , Simulação por Computador , Camundongos
6.
Commun Biol ; 7(1): 550, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719883

RESUMO

Perceptual and cognitive processing relies on flexible communication among cortical areas; however, the underlying neural mechanism remains unclear. Here we report a mechanism based on the realistic spatiotemporal dynamics of propagating wave patterns in neural population activity. Using a biophysically plausible, multiarea spiking neural circuit model, we demonstrate that these wave patterns, characterized by their rich and complex dynamics, can account for a wide variety of empirically observed neural processes. The coordinated interactions of these wave patterns give rise to distributed and dynamic communication (DDC) that enables flexible and rapid routing of neural activity across cortical areas. We elucidate how DDC unifies the previously proposed oscillation synchronization-based and subspace-based views of interareal communication, offering experimentally testable predictions that we validate through the analysis of Allen Institute Neuropixels data. Furthermore, we demonstrate that DDC can be effectively modulated during attention tasks through the interplay of neuromodulators and cortical feedback loops. This modulation process explains many neural effects of attention, underscoring the fundamental functional role of DDC in cognition.


Assuntos
Atenção , Modelos Neurológicos , Atenção/fisiologia , Humanos , Córtex Cerebral/fisiologia , Animais , Rede Nervosa/fisiologia , Percepção Visual/fisiologia , Neurônios/fisiologia , Cognição/fisiologia
7.
Sci Rep ; 14(1): 10536, 2024 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719897

RESUMO

Precisely timed and reliably emitted spikes are hypothesized to serve multiple functions, including improving the accuracy and reproducibility of encoding stimuli, memories, or behaviours across trials. When these spikes occur as a repeating sequence, they can be used to encode and decode a potential time series. Here, we show both analytically and in simulations that the error incurred in approximating a time series with precisely timed and reliably emitted spikes decreases linearly with the number of neurons or spikes used in the decoding. This was verified numerically with synthetically generated patterns of spikes. Further, we found that if spikes were imprecise in their timing, or unreliable in their emission, the error incurred in decoding with these spikes would be sub-linear. However, if the spike precision or spike reliability increased with network size, the error incurred in decoding a time-series with sequences of spikes would maintain a linear decrease with network size. The spike precision had to increase linearly with network size, while the probability of spike failure had to decrease with the square-root of the network size. Finally, we identified a candidate circuit to test this scaling relationship: the repeating sequences of spikes with sub-millisecond precision in area HVC (proper name) of the zebra finch. This scaling relationship can be tested using both neural data and song-spectrogram-based recordings while taking advantage of the natural fluctuation in HVC network size due to neurogenesis.


Assuntos
Potenciais de Ação , Modelos Neurológicos , Neurônios , Animais , Potenciais de Ação/fisiologia , Neurônios/fisiologia , Vocalização Animal/fisiologia , Reprodutibilidade dos Testes
8.
Int J Biol Sci ; 20(7): 2476-2490, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38725863

RESUMO

Peristaltic movements in gut are essential to propel ingested materials through the gastrointestinal tract. Intestinal resident macrophages play an important role in this physiological function through protecting enteric neurons. However, it is incompletely clear how individuals maintain the homeostasis of gut motility. Here we found that NLRP3 is a critical factor in controlling loss of muscularis resident macrophages (MMs), and demonstrate that MMs are involved in the homeostasis of excitatory neurons such as choline acetyltransferase (ChAT)+ and vesicular glutamate transporter 2 (VGLUT2)+ but not inhibitory neuronal nitric oxide synthase (nNOS)+ neurons. NLRP3 knockout (KO) mice had enhanced gut motility and increased neurons, especially excitatory ChAT+ and VGLUT2+ neurons. Single cell analyses showed that there had increased resident macrophages, especially MMs in NLRP3 KO mice. The MM proportion in the resident macrophages was markedly higher than those in wild-type (WT) or caspase 1/11 KO mice. Deletion of the MMs and transplantation of the NLRP3 KO bone marrow cells showed that survival of the gut excitatory ChAT+ and VGLUT2+ neurons was dependent on the MMs. Gut microbiota metabolites ß-hydroxybutyrate (BHB) could promote gut motility through protecting MMs from pyroptosis. Thus, our data suggest that MMs regulated by NLRP3 maintain the homeostasis of excitatory neurons.


Assuntos
Homeostase , Macrófagos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neurônios , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Camundongos , Macrófagos/metabolismo , Neurônios/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Colina O-Acetiltransferase/metabolismo , Colina O-Acetiltransferase/genética , Motilidade Gastrointestinal/fisiologia , Microbioma Gastrointestinal/fisiologia
9.
Cell ; 187(10): 2574-2594.e23, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38729112

RESUMO

High-resolution electron microscopy of nervous systems has enabled the reconstruction of synaptic connectomes. However, we do not know the synaptic sign for each connection (i.e., whether a connection is excitatory or inhibitory), which is implied by the released transmitter. We demonstrate that artificial neural networks can predict transmitter types for presynapses from electron micrographs: a network trained to predict six transmitters (acetylcholine, glutamate, GABA, serotonin, dopamine, octopamine) achieves an accuracy of 87% for individual synapses, 94% for neurons, and 91% for known cell types across a D. melanogaster whole brain. We visualize the ultrastructural features used for prediction, discovering subtle but significant differences between transmitter phenotypes. We also analyze transmitter distributions across the brain and find that neurons that develop together largely express only one fast-acting transmitter (acetylcholine, glutamate, or GABA). We hope that our publicly available predictions act as an accelerant for neuroscientific hypothesis generation for the fly.


Assuntos
Encéfalo , Drosophila melanogaster , Microscopia Eletrônica , Neurônios , Neurotransmissores , Sinapses , Animais , Drosophila melanogaster/ultraestrutura , Drosophila melanogaster/metabolismo , Neurotransmissores/metabolismo , Sinapses/ultraestrutura , Sinapses/metabolismo , Microscopia Eletrônica/métodos , Encéfalo/ultraestrutura , Encéfalo/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Redes Neurais de Computação , Conectoma , Ácido gama-Aminobutírico/metabolismo
10.
Sci Adv ; 10(19): eadk7636, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38728397

RESUMO

Corticotropin releasing factor (CRF) network in the oval nucleus of bed nuclei of the stria terminalis (ovBNST) is generally indicated in stress, but its role in female-biased susceptibility to anxiety is unknown. Here, we established a female-biased stress paradigm. We found that the CRF release in ovBNST during stress showed female-biased pattern, and ovBNST CRF neurons were more prone to be hyperexcited in female mice during stress in both in vitro and in vivo studies. Moreover, optogenetic modulation to exchange the activation pattern of ovBNST CRF neurons during stress between female and male mice could reverse their susceptibility to anxiety. Last, CRF receptor type 1 (CRFR1) mediated the CRF-induced excitation of ovBNST CRF neurons and showed female-biased expression. Specific knockdown of the CRFR1 level in ovBNST CRF neurons in female or overexpression that in male could reverse their susceptibility to anxiety. Therefore, we identify that CRFR1-mediated hyperexcitation of ovBNST CRF neurons in female mice encode the female-biased susceptibility to anxiety.


Assuntos
Ansiedade , Hormônio Liberador da Corticotropina , Neurônios , Receptores de Hormônio Liberador da Corticotropina , Núcleos Septais , Animais , Feminino , Ansiedade/metabolismo , Masculino , Neurônios/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Núcleos Septais/metabolismo , Camundongos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Aprendizagem da Esquiva/fisiologia , Estresse Psicológico/metabolismo , Comportamento Animal
11.
Mol Biol Rep ; 51(1): 649, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38733445

RESUMO

Molecular pathways involved in cerebral stroke are diverse. The major pathophysiological events that are observed in stroke comprises of excitotoxicity, oxidative stress, mitochondrial damage, endoplasmic reticulum stress, cellular acidosis, blood-brain barrier disruption, neuronal swelling and neuronal network mutilation. Various biomolecules are involved in these pathways and several major proteins are upregulated and/or suppressed following stroke. Different types of receptors, ion channels and transporters are activated. Fluctuations in levels of various ions and neurotransmitters have been observed. Cells involved in immune responses and various mediators involved in neuro-inflammation get upregulated progressing the pathogenesis of the disease. Despite of enormity of the problem, there is not a single therapy that can limit infarction and neurological disability due to stroke. This is because of poor understanding of the complex interplay between these pathophysiological processes. This review focuses upon the past to present research on pathophysiological events that are involved in stroke and various factors that are leading to neuronal death following cerebral stroke. This will pave a way to researchers for developing new potent therapeutics that can aid in the treatment of cerebral stroke.


Assuntos
Estresse Oxidativo , Acidente Vascular Cerebral , Humanos , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Animais , Estresse do Retículo Endoplasmático , Neurônios/metabolismo , Neurônios/patologia , Barreira Hematoencefálica/metabolismo , Mitocôndrias/metabolismo
12.
J Comp Neurol ; 532(5): e25620, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38733146

RESUMO

We used diverse methods to characterize the role of avian lateral spiriform nucleus (SpL) in basal ganglia motor function. Connectivity analysis showed that SpL receives input from globus pallidus (GP), and the intrapeduncular nucleus (INP) located ventromedial to GP, whose neurons express numerous striatal markers. SpL-projecting GP neurons were large and aspiny, while SpL-projecting INP neurons were medium sized and spiny. Connectivity analysis further showed that SpL receives inputs from subthalamic nucleus (STN) and substantia nigra pars reticulata (SNr), and that the SNr also receives inputs from GP, INP, and STN. Neurochemical analysis showed that SpL neurons express ENK, GAD, and a variety of pallidal neuron markers, and receive GABAergic terminals, some of which also contain DARPP32, consistent with GP pallidal and INP striatal inputs. Connectivity and neurochemical analysis showed that the SpL input to tectum prominently ends on GABAA receptor-enriched tectobulbar neurons. Behavioral studies showed that lesions of SpL impair visuomotor behaviors involving tracking and pecking moving targets. Our results suggest that SpL modulates brainstem-projecting tectobulbar neurons in a manner comparable to the demonstrated influence of GP internus on motor thalamus and of SNr on tectobulbar neurons in mammals. Given published data in amphibians and reptiles, it seems likely the SpL circuit represents a major direct pathway-type circuit by which the basal ganglia exerts its motor influence in nonmammalian tetrapods. The present studies also show that avian striatum is divided into three spatially segregated territories with differing connectivity, a medial striato-nigral territory, a dorsolateral striato-GP territory, and the ventrolateral INP motor territory.


Assuntos
Gânglios da Base , Vias Neurais , Animais , Gânglios da Base/metabolismo , Vias Neurais/fisiologia , Vias Neurais/química , Masculino , Neurônios/metabolismo , Globo Pálido/metabolismo , Globo Pálido/química , Globo Pálido/anatomia & histologia
13.
Brief Bioinform ; 25(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38739758

RESUMO

The complicated process of neuronal development is initiated early in life, with the genetic mechanisms governing this process yet to be fully elucidated. Single-cell RNA sequencing (scRNA-seq) is a potent instrument for pinpointing biomarkers that exhibit differential expression across various cell types and developmental stages. By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal development. Our focus extends beyond simply identifying DEGs. We strive to investigate the functional roles of these genes through enrichment analysis and construct gene regulatory networks to understand their interactions. Ultimately, this comprehensive approach aspires to illuminate the molecular mechanisms and transcriptional dynamics governing early human brain development. By uncovering potential links between these DEGs and intelligence, mental disorders, and neurodevelopmental disorders, we hope to shed light on human neurological health and disease. In this study, we have used scRNA-seq to identify DEGs involved in early-stage neuronal development in hESCs. The scRNA-seq data, collected on days 26 (D26) and 54 (D54), of the in vitro differentiation of hESCs to neurons were analyzed. Our analysis identified 539 DEGs between D26 and D54. Functional enrichment of those DEG biomarkers indicated that the up-regulated DEGs participated in neurogenesis, while the down-regulated DEGs were linked to synapse regulation. The Reactome pathway analysis revealed that down-regulated DEGs were involved in the interactions between proteins located in synapse pathways. We also discovered interactions between DEGs and miRNA, transcriptional factors (TFs) and DEGs, and between TF and miRNA. Our study identified 20 significant transcription factors, shedding light on early brain development genetics. The identified DEGs and gene regulatory networks are valuable resources for future research into human brain development and neurodevelopmental disorders.


Assuntos
Biomarcadores , Encéfalo , Redes Reguladoras de Genes , Células-Tronco Embrionárias Humanas , Análise de Célula Única , Humanos , Análise de Célula Única/métodos , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Encéfalo/metabolismo , Encéfalo/embriologia , Encéfalo/citologia , Biomarcadores/metabolismo , Neurônios/metabolismo , Neurônios/citologia , Diferenciação Celular/genética , RNA-Seq , Neurogênese/genética , Regulação da Expressão Gênica no Desenvolvimento , Perfilação da Expressão Gênica , Análise de Sequência de RNA/métodos , Análise da Expressão Gênica de Célula Única
14.
J Math Biol ; 89(1): 3, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38740613

RESUMO

Dynamical systems on networks typically involve several dynamical processes evolving at different timescales. For instance, in Alzheimer's disease, the spread of toxic protein throughout the brain not only disrupts neuronal activity but is also influenced by neuronal activity itself, establishing a feedback loop between the fast neuronal activity and the slow protein spreading. Motivated by the case of Alzheimer's disease, we study the multiple-timescale dynamics of a heterodimer spreading process on an adaptive network of Kuramoto oscillators. Using a minimal two-node model, we establish that heterogeneous oscillatory activity facilitates toxic outbreaks and induces symmetry breaking in the spreading patterns. We then extend the model formulation to larger networks and perform numerical simulations of the slow-fast dynamics on common network motifs and on the brain connectome. The simulations corroborate the findings from the minimal model, underscoring the significance of multiple-timescale dynamics in the modeling of neurodegenerative diseases.


Assuntos
Doença de Alzheimer , Encéfalo , Simulação por Computador , Conceitos Matemáticos , Modelos Neurológicos , Neurônios , Humanos , Doença de Alzheimer/fisiopatologia , Neurônios/fisiologia , Encéfalo/fisiopatologia , Conectoma , Doenças Neurodegenerativas/fisiopatologia , Doenças Neurodegenerativas/patologia , Rede Nervosa/fisiopatologia , Rede Nervosa/fisiologia
15.
PLoS Comput Biol ; 20(5): e1012074, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38696532

RESUMO

We investigate the ability of the pairwise maximum entropy (PME) model to describe the spiking activity of large populations of neurons recorded from the visual, auditory, motor, and somatosensory cortices. To quantify this performance, we use (1) Kullback-Leibler (KL) divergences, (2) the extent to which the pairwise model predicts third-order correlations, and (3) its ability to predict the probability that multiple neurons are simultaneously active. We compare these with the performance of a model with independent neurons and study the relationship between the different performance measures, while varying the population size, mean firing rate of the chosen population, and the bin size used for binarizing the data. We confirm the previously reported excellent performance of the PME model for small population sizes N < 20. But we also find that larger mean firing rates and bin sizes generally decreases performance. The performance for larger populations were generally not as good. For large populations, pairwise models may be good in terms of predicting third-order correlations and the probability of multiple neurons being active, but still significantly worse than small populations in terms of their improvement over the independent model in KL-divergence. We show that these results are independent of the cortical area and of whether approximate methods or Boltzmann learning are used for inferring the pairwise couplings. We compared the scaling of the inferred couplings with N and find it to be well explained by the Sherrington-Kirkpatrick (SK) model, whose strong coupling regime shows a complex phase with many metastable states. We find that, up to the maximum population size studied here, the fitted PME model remains outside its complex phase. However, the standard deviation of the couplings compared to their mean increases, and the model gets closer to the boundary of the complex phase as the population size grows.


Assuntos
Entropia , Modelos Neurológicos , Neurônios , Animais , Neurônios/fisiologia , Córtex Cerebral/fisiologia , Potenciais de Ação/fisiologia , Biologia Computacional , Simulação por Computador
16.
Lab Chip ; 24(10): 2811-2824, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38700452

RESUMO

The aging process has broad physiological impacts, including a significant decline in sensory function, which threatens both physical health and quality of life. One ideal model to study aging, neuronal function, and gene expression is the nematode Caenorhabditis elegans, which has a short lifespan and relatively simple, thoroughly mapped nervous system and genome. Previous works have identified that mechanosensory neuronal structure changes with age, but importantly, the actual age-related changes in the function and health of neurons, as well as the underlying genetic mechanisms responsible for these declines, are not fully understood. While advanced techniques such as single-cell RNA-sequencing have been developed to quantify gene expression, it is difficult to relate this information to functional changes in aging due to a lack of tools available. To address these limitations, we present a platform capable of measuring both physiological function and its associated gene expression throughout the aging process in individuals. Using our pipeline, we investigate the age-related changes in function of the mechanosensing ALM neuron in C. elegans, as well as some relevant gene expression patterns (mec-4 and mec-10). Using a series of devices for animals of different ages, we examined subtle changes in neuronal function and found that while the magnitude of neuronal response to a large stimulus declines with age, sensory capability does not significantly decline with age; further, gene expression is well maintained throughout aging. Additionally, we examine PVD, a harsh-touch mechanosensory neuron, and find that it exhibits a similar age-related decline in magnitude of neuronal response. Together, our data demonstrate that our strategy is useful for identifying genetic factors involved in the decline in neuronal health. We envision that this framework could be applied to other systems as a useful tool for discovering new biology.


Assuntos
Envelhecimento , Caenorhabditis elegans , Dispositivos Lab-On-A-Chip , Neurônios , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Envelhecimento/fisiologia , Neurônios/metabolismo , Neurônios/citologia , Mecanotransdução Celular , Técnicas Analíticas Microfluídicas/instrumentação
17.
Biochem Biophys Res Commun ; 716: 150010, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38704892

RESUMO

Calcium (Ca2+) in mitochondria plays crucial roles in neurons including modulating metabolic processes. Moreover, excessive Ca2+ in mitochondria can lead to cell death. Thus, altered mitochondrial Ca2+ regulation has been implicated in several neurodegenerative diseases including Huntington's disease (HD). HD is a progressive hereditary neurodegenerative disorder that results from abnormally expanded cytosine-adenine-guanine trinucleotide repeats in the huntingtin gene. One neuropathological hallmark of HD is neuronal loss in the striatum and cortex. However, mechanisms underlying selective loss of striatal and cortical neurons in HD remain elusive. Here, we measured the basal Ca2+ levels and Ca2+ uptake in single presynaptic mitochondria during 100 external electrical stimuli using highly sensitive mitochondria-targeted Ca2+ indicators in cultured cortical and striatal neurons of a knock-in mouse model of HD (zQ175 mice). We observed elevated presynaptic mitochondrial Ca2+ uptake during 100 electrical stimuli in HD cortical neurons compared with wild-type (WT) cortical neurons. We also found the highly elevated presynaptic mitochondrial basal Ca2+ level and Ca2+ uptake during 100 stimuli in HD striatal neurons. The elevated presynaptic mitochondrial basal Ca2+ level in HD striatal neurons and Ca2+ uptake during stimulation in HD striatal and cortical neurons can disrupt neurotransmission and induce mitochondrial Ca2+ overload, eventually leading to neuronal death in the striatum and cortex of HD.


Assuntos
Cálcio , Córtex Cerebral , Corpo Estriado , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Doença de Huntington , Mitocôndrias , Terminações Pré-Sinápticas , Animais , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Doença de Huntington/genética , Cálcio/metabolismo , Mitocôndrias/metabolismo , Camundongos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Terminações Pré-Sinápticas/metabolismo , Células Cultivadas , Neurônios/metabolismo , Neurônios/patologia , Camundongos Transgênicos
18.
J Cell Biol ; 223(7)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38722279

RESUMO

In addition to its well-established role in actin assembly, profilin 1 (PFN1) has been shown to bind to tubulin and alter microtubule growth. However, whether PFN1's predominant control over microtubules in cells occurs through direct regulation of tubulin or indirectly through the polymerization of actin has yet to be determined. Here, we manipulated PFN1 expression, actin filament assembly, and actomyosin contractility and showed that reducing any of these parameters for extended periods of time caused an adaptive response in the microtubule cytoskeleton, with the effect being significantly more pronounced in neuronal processes. All the observed changes to microtubules were reversible if actomyosin was restored, arguing that PFN1's regulation of microtubules occurs principally through actin. Moreover, the cytoskeletal modifications resulting from PFN1 depletion in neuronal processes affected microtubule-based transport and mimicked phenotypes that are linked to neurodegenerative disease. This demonstrates how defects in actin can cause compensatory responses in other cytoskeleton components, which in turn significantly alter cellular function.


Assuntos
Actinas , Microtúbulos , Profilinas , Animais , Humanos , Camundongos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actinas/genética , Actomiosina/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Profilinas/metabolismo , Profilinas/genética , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética
19.
J Nanobiotechnology ; 22(1): 260, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38760847

RESUMO

Neurodegenerative diseases are characterized by extensive loss of function or death of brain cells, hampering the life quality of patients. Brain-targeted drug delivery is challenging, with a low success rate this far. Therefore, the application of targeting ligands in drug vehicles, such as lipid-based and polymeric nanoparticles, holds the promise to overcome the blood-brain barrier (BBB) and direct therapies to the brain, in addition to protect their cargo from degradation and metabolization. In this review, we discuss the barriers to brain delivery and the different types of brain-targeting ligands currently in use in brain-targeted nanoparticles, such as peptides, proteins, aptamers, small molecules, and antibodies. Moreover, we present a detailed review of the different targeting ligands used to direct nanoparticles to specific brain cells, like neurons (C4-3 aptamer, neurotensin, Tet-1, RVG, and IKRG peptides), astrocytes (Aquaporin-4, D4, and Bradykinin B2 antibodies), oligodendrocytes (NG-2 antibody and the biotinylated DNA aptamer conjugated to a streptavidin core Myaptavin-3064), microglia (CD11b antibody), neural stem cells (QTRFLLH, VPTQSSG, and NFL-TBS.40-63 peptides), and to endothelial cells of the BBB (transferrin and insulin proteins, and choline). Reports demonstrated enhanced brain-targeted delivery with improved transport to the specific cell type targeted with the conjugation of these ligands to nanoparticles. Hence, this strategy allows the implementation of high-precision medicine, with reduced side effects or unwanted therapy clearance from the body. Nevertheless, the accumulation of some of these nanoparticles in peripheral organs has been reported indicating that there are still factors to be improved to achieve higher levels of brain targeting. This review is a collection of studies exploring targeting ligands for the delivery of nanoparticles to the brain and we highlight the advantages and limitations of this type of approach in precision therapies.


Assuntos
Barreira Hematoencefálica , Encéfalo , Sistemas de Liberação de Medicamentos , Nanopartículas , Humanos , Barreira Hematoencefálica/metabolismo , Animais , Encéfalo/metabolismo , Ligantes , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Peptídeos/química
20.
Zhen Ci Yan Jiu ; 49(5): 480-486, 2024 May 25.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38764119

RESUMO

OBJECTIVES: To observe the activation state and neuronal types of somatosensory cortex and the primary motor cortex induced by electroacupuncture (EA) stimulation of "Sibai" (ST2) and "Quanliao" (SI18) acupoints in mice. METHODS: Male C57BL/6J mice were randomly divided into blank control and EA groups, with 6 mice in each group. Rats of the EA group received EA stimulation (2 Hz, 0.6 mA) at ST2 and SI18 for 30 minutes. Samples were collected after EA intervention, and immunofluorescence staining was performed to quantify the expression of the c-Fos gene (proportion of c-Fos positive cells) in the somatosensory cortex and primary motor cortex. The co-labelled cells of calcium/calmodulin-dependent protein kinase Ⅱ (CaMKⅡ) and gamma-aminobutyric acid (GABA) in the somatosensory cortex and primary motor cortex were observed and counted by using microscope after immunofluorescence staining. Another 10 mice were used to detect the calcium activity of excitatory neurons in the somatosensory cortex and primary motor cortex by fiber photometry. RESULTS: In comparison with the blank control group, the number of c-Fos positive cells, and the proportion of c-Fos and CaMKⅡ co-labelled cells in both the somatosensory cortex and primary motor cortex were significantly increased after EA stimulation (P<0.05). No significant changes were found in the proportion of c-Fos and GABA co-labeled cells in both the somatosensory cortex and primary motor cortex after EA. Results of fiber optic calcium imaging technology showed that the spontaneous calcium activity of excitatory neurons in both somatosensory cortex and primary motor cortex were obviously increased during EA compared with that before EA (P<0.01), and strikingly reduced after cessation of EA compared with that during EA (P<0.05). CONCLUSIONS: Under physiological conditions, EA of ST2 and SI18 can effectively activate excitatory neurons in the somatosensory cortex and primary motor cortex.


Assuntos
Pontos de Acupuntura , Eletroacupuntura , Camundongos Endogâmicos C57BL , Neurônios , Animais , Masculino , Camundongos , Neurônios/metabolismo , Córtex Sensório-Motor/metabolismo , Humanos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Córtex Motor/metabolismo , Córtex Somatossensorial/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...