Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.027
Filtrar
1.
Mol Cell ; 84(18): 3530-3544.e6, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39232582

RESUMO

Channelrhodopsins are microbial light-gated ion channels that can control the firing of neurons in response to light. Among several cation channelrhodopsins identified in Guillardia theta (GtCCRs), GtCCR4 has higher light sensitivity than typical channelrhodopsins. Furthermore, GtCCR4 shows superior properties as an optogenetic tool, such as minimal desensitization. Our structural analyses of GtCCR2 and GtCCR4 revealed that GtCCR4 has an outwardly bent transmembrane helix, resembling the conformation of activated G-protein-coupled receptors. Spectroscopic and electrophysiological comparisons suggested that this helix bend in GtCCR4 omits channel recovery time and contributes to high light sensitivity. An electrophysiological comparison of GtCCR4 and the well-characterized optogenetic tool ChRmine demonstrated that GtCCR4 has superior current continuity and action-potential spike generation with less invasiveness in neurons. We also identified highly active mutants of GtCCR4. These results shed light on the diverse structures and dynamics of microbial rhodopsins and demonstrate the strong optogenetic potential of GtCCR4.


Assuntos
Bacteriorodopsinas , Luz , Neurônios , Optogenética , Optogenética/métodos , Animais , Neurônios/metabolismo , Neurônios/efeitos da radiação , Bacteriorodopsinas/metabolismo , Bacteriorodopsinas/genética , Bacteriorodopsinas/química , Humanos , Células HEK293 , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Channelrhodopsins/química , Potenciais de Ação , Criptófitas/genética , Criptófitas/metabolismo , Mutação , Ativação do Canal Iônico/efeitos da radiação , Relação Estrutura-Atividade
2.
J Photochem Photobiol B ; 259: 113021, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39222549

RESUMO

Alzheimer's disease, a prevalent neurodegenerative condition primarily affecting older adults, remains incurable. Its principle pathological hallmark is the accelerated accumulation of amyloid ß (Aß) protein. This study investigates the potential of photobiomodulation using near infrared light to counteract Aß1-42-induced synaptic degeneration and neurotoxicity. We focused on the effect of 808 nm near-infrared laser diode (LD) on Aß1-42 cytotoxicity in primary cultured cortical neurons. We assessed cell survival using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, observing substantial benefits from LD irradiation with a power of 10 mW and a dose of 30 J. Cells exposed to Aß1-42 exhibited morphological changes indicative of synaptic damage and a significant decrease in the number of postsynaptic density protein-95 (PSD-95) contacts, which were significantly improved with near-infrared LD therapy. Furthermore, this therapy reduced Aß and phosphorylated tau (P-tau) protein accumulation. Additionally, near-infrared LD irradiation substantially lessened the Aß1-42-induced rise in glial fibrillary acid protein (GFAP) and ionized calcium-binding adaptor molecule 1 (IBA1) in astrocytes and microglia. Remarkably, near-infrared LD irradiation effectively inhibited phosphorylation of key proteins involved in Aß1-42-induced necroptosis, namely Receptor-interacting protein kinase-3 (RIP3) and Mixed Lineage Kinase domain-Like protein (MLKL). Our findings suggest that near-infrared LD treatment significantly reduces neurodegeneration by reducing glial overactivation and neuronal necroptosis triggered by Aß1-42. Thus, near-infrared LD treatment emerges as a promising approach for slowing or treating Alzheimer's disease, offering new avenues in its management.


Assuntos
Peptídeos beta-Amiloides , Sobrevivência Celular , Raios Infravermelhos , Neurônios , Fragmentos de Peptídeos , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Neurônios/efeitos da radiação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Ratos , Lasers Semicondutores , Proteínas tau/metabolismo , Terapia com Luz de Baixa Intensidade , Células Cultivadas , Proteína 4 Homóloga a Disks-Large/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/efeitos da radiação , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/efeitos da radiação
3.
Sci Rep ; 14(1): 21107, 2024 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-39256554

RESUMO

Different wavelengths emitted from light-emitting diodes (LEDs) are known as an influential factor in proliferation and differentiation of various cell types. Since human umbilical cord matrix-derived mesenchymal cells (hUCMs) are ideal tools for human regenerative medicine clinical trials and stem cell researches, in the present study we investigated the neurogenesis effects of single and intermittent green and red LED irradiation on hUCM cells. Exposure of hUCMs to single and intermittent green (530 nm, 1.59 J/cm2) and red (630 nm, 0.318 J/cm2) lights significantly increased the expression of specific genes including nestin, ß-tubulin III and Olig2. Additionally, immunocytochemical analysis confirmed the expression of specific neural-related proteins including nestin, ß-tubulin III, Olig2 and GFAP. Also, alternating exposure of hUCM cells to green and red lights increased the expression of some neural markers more than either light alone. Further research are required to develop the application of LED irradiation as a useful tool for therapeutic purposes including neural repair and regeneration.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais , Neurogênese , Cordão Umbilical , Humanos , Células-Tronco Mesenquimais/efeitos da radiação , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular/efeitos da radiação , Cordão Umbilical/citologia , Neurogênese/efeitos da radiação , Luz , Nestina/metabolismo , Nestina/genética , Células Cultivadas , Neurônios/efeitos da radiação , Neurônios/metabolismo , Neurônios/citologia , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/genética
4.
Nat Commun ; 15(1): 6768, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39117652

RESUMO

Light is fundamental for biological life, with most mammals possessing light-sensing photoreceptors in various organs. Opsin3 is highly expressed in adipose tissue which has extensive communication with other organs, particularly with the brain through the sympathetic nervous system (SNS). Our study reveals a new light-triggered crosstalk between adipose tissue and the hypothalamus. Direct blue-light exposure to subcutaneous white fat improves high-fat diet-induced metabolic abnormalities in an Opsin3-dependent manner. Metabolomic analysis shows that blue light increases circulating levels of histidine, which activates histaminergic neurons in the hypothalamus and stimulates brown adipose tissue (BAT) via SNS. Blocking central actions of histidine and denervating peripheral BAT blunts the effects of blue light. Human white adipocytes respond to direct blue light stimulation in a cell-autonomous manner, highlighting the translational relevance of this pathway. Together, these data demonstrate a light-responsive metabolic circuit involving adipose-hypothalamus communication, offering a potential strategy to alleviate obesity-induced metabolic abnormalities.


Assuntos
Tecido Adiposo Marrom , Hipotálamo , Luz , Animais , Hipotálamo/metabolismo , Hipotálamo/efeitos da radiação , Humanos , Tecido Adiposo Marrom/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Camundongos Endogâmicos C57BL , Dieta Hiperlipídica/efeitos adversos , Opsinas de Bastonetes/metabolismo , Sistema Nervoso Simpático/metabolismo , Tecido Adiposo/metabolismo , Neurônios/metabolismo , Neurônios/efeitos da radiação , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/efeitos da radiação , Adipócitos Brancos/metabolismo , Adipócitos Brancos/efeitos da radiação
5.
J Photochem Photobiol B ; 258: 112998, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39096719

RESUMO

Depression, a multifactorial mental disorder, characterized by cognitive slowing, anxiety, and impaired cognitive function, imposes a significant burden on public health. Photobiomodulation (PBM), involving exposure to sunlight or artificial light at a specific intensity and wavelength for a determined duration, influences brain activity, functional connectivity, and plasticity. It is recognized for its therapeutic efficacy in treating depression, yet its molecular and cellular underpinnings remain obscure. Here, we investigated the impact of PBM with 468 nm light on depression-like behavior and neuronal damage in the chronic unpredictable mild stress (CUMS) murine model, a commonly employed animal model for studying depression. Our results demonstrate that PBM treatment ameliorated behavioral deficits, inhibited neuroinflammation and apoptosis, and notably rejuvenates the hippocampal synaptic function in depressed mice, which may be mainly attributed to the up-regulation of brain-derived neurotrophic factor signaling pathways. In addition, in vitro experiments with a corticosterone-induced hippocampal neuron injury model demonstrate reduced oxidative stress and improved mitochondrial function, further validating the therapeutic potential of PBM. In summary, these findings suggest PBM as a promising, non-invasive treatment for depression, offering insights into its biological mechanisms and potential for clinical application.


Assuntos
Depressão , Modelos Animais de Doenças , Hipocampo , Terapia com Luz de Baixa Intensidade , Mitocôndrias , Animais , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Camundongos , Depressão/metabolismo , Depressão/terapia , Hipocampo/efeitos da radiação , Hipocampo/metabolismo , Masculino , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sinapses/efeitos da radiação , Sinapses/metabolismo , Estresse Oxidativo/efeitos da radiação , Camundongos Endogâmicos C57BL , Neurônios/efeitos da radiação , Neurônios/metabolismo , Plasticidade Neuronal/efeitos da radiação , Corticosterona , Comportamento Animal/efeitos da radiação , Apoptose/efeitos da radiação , Estresse Psicológico
7.
Sci Rep ; 14(1): 20143, 2024 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-39210021

RESUMO

Radiation therapy and stereotactic radiosurgery are common treatments for brain malignancies. However, the impact of radiation on underlying neuronal circuits is poorly understood. In the prefrontal cortex (PFC), neurons communicate via action potentials that control cognitive processes, thus it is important to understand the impact of radiation on these circuits. Here we present a novel protocol to investigate the effect of radiation on the activity and survival of PFC networks in vitro. Escalating doses of radiation were applied to PFC slices using a robotic radiosurgery platform at a standard dose rate of 10 Gy/min. High-density multielectrode array recordings of radiated slices were collected to capture extracellular activity across 4,096 channels. Radiated slices showed an increase in firing rate, functional connectivity, and complexity. Graph-theoretic measures of functional connectivity were altered following radiation. These results were compared to pharmacologically induced epileptic slices where neural complexity was markedly elevated, and functional connections were strong but remained spatially focused. Finally, propidium iodide staining revealed a dose-dependent effect of radiation on apoptosis. These findings provide a novel assay to investigate the impacts of clinically relevant doses of radiation on brain circuits and highlight the acute effects of escalating radiation doses on PFC neurons.


Assuntos
Potenciais de Ação , Neurônios , Córtex Pré-Frontal , Animais , Córtex Pré-Frontal/efeitos da radiação , Córtex Pré-Frontal/fisiologia , Neurônios/efeitos da radiação , Neurônios/fisiologia , Potenciais de Ação/efeitos da radiação , Rede Nervosa/efeitos da radiação , Rede Nervosa/fisiologia , Masculino , Eletrodos , Ratos , Apoptose/efeitos da radiação , Radiocirurgia/métodos
8.
PLoS One ; 19(8): e0306656, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39213294

RESUMO

The invention of Light Emitting Diode (LED) revolutionized energy-efficient illumination, but concerns persist regarding the potential harm of blue light to our eyes. In this study, we scrutinized the impact of LED light characteristics on eyes using two cell types: M-1 (rich in mitochondria) and CD-1 (neuronal). Variations in color rendering index (CRI) and correlated color temperature (CCT) were investigated, alongside exposure durations ranging from 0 to 24 hours. The findings illuminated the potential benefits of high-quality LED lighting, characterized by a high CRI and low CCT, which emits a greater proportion of red light. This form of lighting was associated with enhanced cell proliferation, elevated ATP levels, and reduced oxidative stress. In contrast, LEDs with low CRI and high CCT exhibited adverse effects, diminishing cell viability and increasing oxidative stress. These results suggest that high-quality LED lighting may have neuroprotective potential as a treatment option, such as for retinal ganglion cells.


Assuntos
Sobrevivência Celular , Luz , Mitocôndrias , Neurônios , Estresse Oxidativo , Animais , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Neurônios/metabolismo , Neurônios/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Córtex Cerebral/efeitos da radiação , Córtex Cerebral/metabolismo , Córtex Cerebral/citologia , Linhagem Celular , Proliferação de Células/efeitos da radiação , Trifosfato de Adenosina/metabolismo , Iluminação
9.
Nature ; 633(8030): 615-623, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39169183

RESUMO

Mosquito-borne diseases affect hundreds of millions of people annually and disproportionately impact the developing world1,2. One mosquito species, Aedes aegypti, is a primary vector of viruses that cause dengue, yellow fever and Zika. The attraction of Ae. aegypti female mosquitos to humans requires integrating multiple cues, including CO2 from breath, organic odours from skin and visual cues, all sensed at mid and long ranges, and other cues sensed at very close range3-6. Here we identify a cue that Ae. aegypti use as part of their sensory arsenal to find humans. We demonstrate that Ae. aegypti sense the infrared (IR) radiation emanating from their targets and use this information in combination with other cues for highly effective mid-range navigation. Detection of thermal IR requires the heat-activated channel TRPA1, which is expressed in neurons at the tip of the antenna. Two opsins are co-expressed with TRPA1 in these neurons and promote the detection of lower IR intensities. We propose that radiant energy causes local heating at the end of the antenna, thereby activating temperature-sensitive receptors in thermosensory neurons. The realization that thermal IR radiation is an outstanding mid-range directional cue expands our understanding as to how mosquitoes are exquisitely effective in locating hosts.


Assuntos
Aedes , Sinais (Psicologia) , Comportamento de Busca por Hospedeiro , Temperatura Alta , Raios Infravermelhos , Navegação Espacial , Sensação Térmica , Animais , Feminino , Humanos , Aedes/citologia , Aedes/fisiologia , Aedes/efeitos da radiação , Antenas de Artrópodes/citologia , Antenas de Artrópodes/inervação , Antenas de Artrópodes/fisiologia , Comportamento de Busca por Hospedeiro/fisiologia , Comportamento de Busca por Hospedeiro/efeitos da radiação , Mosquitos Vetores/citologia , Mosquitos Vetores/fisiologia , Mosquitos Vetores/efeitos da radiação , Neurônios/efeitos da radiação , Neurônios/metabolismo , Neurônios/fisiologia , Opsinas/metabolismo , Sensação Térmica/fisiologia , Sensação Térmica/efeitos da radiação , Canal de Cátion TRPA1/metabolismo , Dióxido de Carbono/metabolismo , Odor Corporal , Navegação Espacial/fisiologia , Navegação Espacial/efeitos da radiação
10.
Int J Mol Sci ; 25(16)2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39201275

RESUMO

The widespread use of wireless communication devices has necessitated unavoidable exposure to radiofrequency electromagnetic fields (RF-EMF). In particular, increasing RF-EMF exposure among children is primarily driven by mobile phone use. Therefore, this study investigated the effects of 1850 MHz RF-EMF exposure at a specific absorption rate of 4.0 W/kg on cortical neurons in mice at postnatal day 28. The results indicated a significant reduction in the number of mushroom-shaped dendritic spines in the prefrontal cortex after daily exposure for 4 weeks. Additionally, prolonged RF-EMF exposure over 9 days led to a gradual decrease in postsynaptic density 95 puncta and inhibited neurite outgrowth in developing cortical neurons. Moreover, the expression levels of genes associated with synapse formation, such as synaptic cell adhesion molecules and cyclin-dependent kinase 5, were reduced in the cerebral cortexes of RF-EMF-exposed mice. Behavioral assessments using the Morris water maze revealed altered spatial learning and memory after the 4-week exposure period. These findings underscore the potential of RF-EMF exposure during childhood to disrupt synaptic function in the cerebral cortex, thereby affecting the developmental stages of the nervous system and potentially influencing later cognitive function.


Assuntos
Neurônios , Ondas de Rádio , Sinapses , Animais , Camundongos , Sinapses/efeitos da radiação , Sinapses/metabolismo , Neurônios/efeitos da radiação , Neurônios/metabolismo , Ondas de Rádio/efeitos adversos , Campos Eletromagnéticos/efeitos adversos , Córtex Cerebral/efeitos da radiação , Córtex Cerebral/metabolismo , Espinhas Dendríticas/efeitos da radiação , Espinhas Dendríticas/metabolismo , Memória/efeitos da radiação , Aprendizagem em Labirinto/efeitos da radiação , Masculino , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 5 Dependente de Ciclina/genética , Crescimento Neuronal/efeitos da radiação , Aprendizagem/efeitos da radiação , Córtex Pré-Frontal/efeitos da radiação , Córtex Pré-Frontal/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo
11.
ACS Chem Neurosci ; 15(17): 3106-3112, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39163542

RESUMO

Dendritic spines function as postsynaptic sites, receiving excitatory signals from presynaptic axons. The synaptic plasticity of spines underlies the refinement of neuronal circuits. Neural cognitive disorders are commonly associated with the impairment and elimination of dendritic spines. In this study, we report an all-optical method to activate dendritic spine growth and regeneration by a single short flash of femtosecond laser stimulation. The inhibited development and loss of spines can be rescued by a transient illumination of the laser inside a micrometer region of the soma by activating the extracellular signal-regulated kinase (ERK) signaling pathway. The rescued neurons exhibit function. Hence we provide a potential noninvasive method for the regeneration of dendritic spines.


Assuntos
Espinhas Dendríticas , Lasers , Animais , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/efeitos da radiação , Neurogênese/fisiologia , Neurogênese/efeitos da radiação , Neurônios/efeitos da radiação , Neurônios/fisiologia , Regeneração Nervosa/fisiologia , Regeneração Nervosa/efeitos da radiação , Plasticidade Neuronal/fisiologia , Plasticidade Neuronal/efeitos da radiação , Sistema de Sinalização das MAP Quinases/fisiologia , Células Cultivadas
12.
Biointerphases ; 19(4)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-39007691

RESUMO

Retinal degenerative diseases, which can lead to photoreceptor cell apoptosis, have now become the leading irreversible cause of blindness worldwide. In this study, we developed an organic photovoltaic biomaterial for artificial retinas, enabling neural cells to detect photoelectric stimulation. The biomaterial was prepared using a conjugated polymer donor, PCE-10, and a non-fullerene receptor, Y6, both known for their strong near-infrared light absorption capabilities. Additionally, a fullerene receptor, PC61BM, was incorporated, which possesses the ability to absorb reactive oxygen species. We conducted a comprehensive investigation into the microstructure, photovoltaic properties, and photothermal effects of this three-component photovoltaic biomaterial. Furthermore, we employed Rat adrenal pheochromocytoma cells (PC-12) as a standard neural cell model to evaluate the in vitro photoelectric stimulation effect of this photovoltaic biomaterial. The results demonstrate that the photovoltaic biomaterial, enriched with fullerene derivatives, can induce intracellular calcium influx in PC-12 cells under 630 nm (red light) and 780 nm (near-infrared) laser irradiation. Moreover, there were lower levels of oxidative stress and higher levels of mitochondrial activity compared to the non-PC61BM group. This photovoltaic biomaterial proves to be an ideal substrate for near-infrared photoelectrical stimulation of neural cells and holds promise for restoring visual function in patients with photoreceptor apoptosis.


Assuntos
Materiais Biocompatíveis , Fulerenos , Raios Infravermelhos , Animais , Fulerenos/química , Fulerenos/farmacologia , Ratos , Materiais Biocompatíveis/química , Células PC12 , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Cálcio/metabolismo , Cálcio/química
13.
Photobiomodul Photomed Laser Surg ; 42(8): 524-533, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39058735

RESUMO

Objective: To investigate the effects of photobiomodulation therapy (PBMT) at 660 and 810 nm on amyloid-beta (Aß)42-induced toxicity in differentiated SH-SY5Y cells and to assess its impact on Aß42 accumulation and cholinergic neurotransmission. Background: Alzheimer's disease (AD) is characterized by the accumulation of Aß peptides, leading to neurodegeneration, cholinergic deficit, and cognitive decline. PBMT has emerged as a potential therapeutic approach to mitigate Aß-induced toxicity and enhance cholinergic function. Methods: Differentiated neurons were treated with 1 µM Aß42 for 1 day, followed by daily PBMT at wavelengths of 660 and 810 nm for 7 days. Treatments used LEDs emitting continuous wave light at a power density of 5 mW/cm2 for 10 min daily to achieve an energy density of 3 J/cm2. Results: Differentiated SH-SY5Y cells exhibited increased Aß42 aggregation, neurite retraction, and reduced cell viability. PBMT at 810 nm significantly mitigated the Aß42-induced toxicity in these cells, as evidenced by reduced Aß42 aggregation, neurite retraction, and improved cell viability and neuronal morphology. Notably, this treatment also restored acetylcholine levels in the neurons exposed to Aß42. Conclusions: PBMT at 810 nm effectively reduces Aß42-induced toxicity and supports neuronal survival, highlighting its neuroprotective effects on cholinergic neurons. By shedding light on the impact of low-level light therapy on Aß42 accumulation and cellular processes. These findings advocate for further research to elucidate the mechanisms of PBMT and validate its clinical relevance in AD management.


Assuntos
Acetilcolina , Doença de Alzheimer , Peptídeos beta-Amiloides , Sobrevivência Celular , Terapia com Luz de Baixa Intensidade , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/radioterapia , Doença de Alzheimer/metabolismo , Humanos , Acetilcolina/metabolismo , Sobrevivência Celular/efeitos da radiação , Neurônios/efeitos da radiação , Neurônios/metabolismo , Linhagem Celular Tumoral , Fragmentos de Peptídeos/metabolismo
14.
Int J Mol Sci ; 25(14)2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39063113

RESUMO

Exposure to 2.45 GHz electromagnetic radiation (EMR) emitted from commonly used devices has been reported to induce oxidative stress in several experimental models. Our study aims to evaluate the efficacy of sulforaphane, a well-known natural product, in preventing radiation-induced toxic effects caused by a 24 h exposure of SH-SY5Y neuronal-like cells and peripheral blood mononuclear cells (PBMCs) to 2.45 GHz EMR. Cells were exposed to radiation for 24 h in the presence or absence of sulforaphane at different concentrations (5-10-25 µg/mL). Cell viability, mitochondrial activity alterations, the transcription and protein levels of redox markers, and apoptosis-related genes were investigated. Our data showed a reduction in cell viability of both neuronal-like cells and PBMCs caused by EMR exposure and a protective effect of 5 µg/mL sulforaphane. The lowest sulforaphane concentration decreased ROS production and increased the Mitochondrial Transmembrane Potential (Δψm) and the NAD+/NADH ratio, which were altered by radiation exposure. Sulforaphane at higher concentrations displayed harmful effects. The hormetic behavior of sulforaphane was also evident after evaluating the expression of genes coding for Nrf2, SOD2, and changes in apoptosis markers. Our study underlined the vulnerability of neuronal-like cells to mitochondrial dysfunction and oxidative stress and the possibility of mitigating these effects by supplementation with sulforaphane. To our knowledge, there are no previous studies about the effects of SFN on these cells when exposed to 2.45 GHz electromagnetic radiation.


Assuntos
Radiação Eletromagnética , Isotiocianatos , Leucócitos Mononucleares , Potencial da Membrana Mitocondrial , Neurônios , Estresse Oxidativo , Sulfóxidos , Isotiocianatos/farmacologia , Humanos , Sulfóxidos/farmacologia , Leucócitos Mononucleares/efeitos da radiação , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Neurônios/efeitos da radiação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Mitocôndrias/metabolismo , Linhagem Celular Tumoral
15.
Nat Commun ; 15(1): 5501, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38951486

RESUMO

While light can affect emotional and cognitive processes of the medial prefrontal cortex (mPFC), no light-encoding was hitherto identified in this region. Here, extracellular recordings in awake mice revealed that over half of studied mPFC neurons showed photosensitivity, that was diminished by inhibition of intrinsically photosensitive retinal ganglion cells (ipRGCs), or of the upstream thalamic perihabenular nucleus (PHb). In 15% of mPFC photosensitive neurons, firing rate changed monotonically along light-intensity steps and gradients. These light-intensity-encoding neurons comprised four types, two enhancing and two suppressing their firing rate with increased light intensity. Similar types were identified in the PHb, where they exhibited shorter latency and increased sensitivity. Light suppressed prelimbic activity but boosted infralimbic activity, mirroring the regions' contrasting roles in fear-conditioning, drug-seeking, and anxiety. We posit that prefrontal photosensitivity represents a substrate of light-susceptible, mPFC-mediated functions, which could be ultimately studied as a therapeutical target in psychiatric and addiction disorders.


Assuntos
Luz , Camundongos Endogâmicos C57BL , Neurônios , Córtex Pré-Frontal , Células Ganglionares da Retina , Animais , Córtex Pré-Frontal/fisiologia , Córtex Pré-Frontal/efeitos da radiação , Córtex Pré-Frontal/citologia , Camundongos , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/efeitos da radiação , Masculino , Neurônios/fisiologia , Neurônios/metabolismo , Neurônios/efeitos da radiação , Estimulação Luminosa , Potenciais de Ação/fisiologia
16.
J Neural Eng ; 21(3)2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38862011

RESUMO

Objective.Commonly used cable equation approaches for simulating the effects of electromagnetic fields on excitable cells make several simplifying assumptions that could limit their predictive power. Bidomain or 'whole' finite element methods have been developed to fully couple cells and electric fields for more realistic neuron modeling. Here, we introduce a novel bidomain integral equation designed for determining the full electromagnetic coupling between stimulation devices and the intracellular, membrane, and extracellular regions of neurons.Approach.Our proposed boundary element formulation offers a solution to an integral equation that connects the device, tissue inhomogeneity, and cell membrane-induced E-fields. We solve this integral equation using first-order nodal elements and an unconditionally stable Crank-Nicholson time-stepping scheme. To validate and demonstrate our approach, we simulated cylindrical Hodgkin-Huxley axons and spherical cells in multiple brain stimulation scenarios.Main Results.Comparison studies show that a boundary element approach produces accurate results for both electric and magnetic stimulation. Unlike bidomain finite element methods, the bidomain boundary element method does not require volume meshes containing features at multiple scales. As a result, modeling cells, or tightly packed populations of cells, with microscale features embedded in a macroscale head model, is simplified, and the relative placement of devices and cells can be varied without the need to generate a new mesh.Significance.Device-induced electromagnetic fields are commonly used to modulate brain activity for research and therapeutic applications. Bidomain solvers allow for the full incorporation of realistic cell geometries, device E-fields, and neuron populations. Thus, multi-cell studies of advanced neuronal mechanisms would greatly benefit from the development of fast-bidomain solvers to ensure scalability and the practical execution of neural network simulations with realistic neuron morphologies.


Assuntos
Campos Eletromagnéticos , Análise de Elementos Finitos , Modelos Neurológicos , Neurônios , Neurônios/fisiologia , Neurônios/efeitos da radiação , Humanos , Simulação por Computador , Animais , Axônios/fisiologia , Axônios/efeitos da radiação , Potenciais de Ação/fisiologia , Potenciais de Ação/efeitos da radiação , Encéfalo/fisiologia
17.
Phytomedicine ; 132: 155803, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38876008

RESUMO

BACKGROUND: Electromagnetic radiation is relevant to human life, and radiation can trigger neurodegenerative diseases by altering the function of the central nervous system through oxidative stress, mitochondrial dysfunction, and protein degradation. Astragaloside IV (AS-IV) is anti-oxidative, anti-apoptotic, activates the BDNF-TrkB pathway and enhances synaptic plasticity in radiated mice, which can exert its neuroprotection. However, the exact molecular mechanisms are still unclear. PURPOSE: This study investigated whether AS-IV could play a neuroprotective role by regulating BDNF-TrkB pathway in radiation damage and its underlying molecular mechanisms. METHODS: Transgenic mice (Thy1-YFP line H) were injected with AS-IV (40 mg/kg/day body weight) by intraperitoneal injection daily for 4 weeks, followed by X-rays. PC12 cells and primary cortical neurons were also exposed to UVA after 24 h of AS-IV treatment (25 µg/ml and 50 µg/ml) in vitro. The impact of radiation on learning and cognitive functions was visualized in the Morris water maze assay. Subsequently, Immunofluorescence and Golgi-Cox staining analyses were utilized to investigate the structural damage of neuronal dendrites and the density of dendritic spines. Transmission electron microscopy was performed to examine how the radiation affected the ultrastructure of neurons. Finally, western blotting analysis and Quantitative RT-PCR were used to evaluate the expression levels and locations of proteins in vitro and in vivo. RESULTS: Radiation induced BDNF-TrkB signaling dysregulation and decreased the levels of neuron-related functional genes (Ngf, Bdnf, Gap-43, Ras, Psd-95, Arc, Creb, c-Fos), PSD-95 and F-actin, which subsequently led to damage of neuronal ultrastructure and dendrites, loss of dendritic spines, and decreased dendritic complexity index, contributing to spatial learning and memory deficits. These abnormalities were prevented by AS-IV treatment. In addition, TrkB receptor antagonists antagonized these neuroprotective actions of AS-IV. 7,8-dihydroxyflavone and AS-IV had neuroprotective effects after radiation. CONCLUSION: AS-IV inhibits morphological damage of neurons and cognitive dysfunction in mice after radiation exposure, resulting in a neuroprotective effect, which were mediated by activating the BDNF-TrkB pathway.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Camundongos Transgênicos , Neurônios , Fármacos Neuroprotetores , Receptor trkB , Saponinas , Transdução de Sinais , Triterpenos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Saponinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Triterpenos/farmacologia , Camundongos , Receptor trkB/metabolismo , Fármacos Neuroprotetores/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Ratos , Células PC12 , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos da radiação
18.
Neuropathol Appl Neurobiol ; 50(3): e12992, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38831600

RESUMO

PURPOSE: Radiation-induced brain injury, one of the side effects of cranial radiotherapy in tumour patients, usually results in durable and serious cognitive disorders. Microglia are important innate immune-effector cells in the central nervous system. However, the interaction between microglia and neurons in radiation-induced brain injury remains uncharacterised. METHODS AND MATERIALS: We established a microglia-neuron indirect co-culture model to assess the interaction between them. Microglia exposed to radiation were examined for pyroptosis using lactate dehydrogenase (LDH) release, Annexin V/PI staining, SYTOX staining and western blot. The role of nucleotide-binding oligomerisation domain-like receptor family pyrin domain containing 3 (NLRP3) was investigated in microglia exposed to radiation and in mouse radiation brain injury model through siRNA or inhibitor. Mini-mental state examination and cytokines in blood were performed in 23 patients who had experienced cranial irradiation. RESULTS: Microglia exerted neurotoxic features after radiation in the co-culture model. NLRP3 was up-regulated in microglia exposed to radiation, and then caspase-1 was activated. Thus, the gasdermin D protein was cleaved, and it triggered pyroptosis in microglia, which released inflammatory cytokines. Meanwhile, treatment with siRNA NLRP3 in vitro and NLRP3 inhibitor in vivo attenuated the damaged neuron cell and cognitive impairment, respectively. What is more, we found that the patients after radiation with higher IL-6 were observed to have a decreased MMSE score. CONCLUSIONS: These findings indicate that radiation-induced pyroptosis in microglia may promote radiation-induced brain injury via the secretion of neurotoxic cytokines. NLRP3 was evaluated as an important mediator in radiation-induced pyroptosis and a promising therapeutic target for radiation-induced brain injury.


Assuntos
Lesões Encefálicas , Microglia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Piroptose , Piroptose/efeitos da radiação , Piroptose/fisiologia , Microglia/metabolismo , Microglia/efeitos da radiação , Microglia/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Camundongos , Humanos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/etiologia , Masculino , Neurônios/metabolismo , Neurônios/patologia , Neurônios/efeitos da radiação , Técnicas de Cocultura , Lesões por Radiação/patologia , Lesões por Radiação/metabolismo , Feminino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
19.
Cells ; 13(11)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38891031

RESUMO

Apolipoprotein E (ApoE) is a lipid carrier in both the peripheral and the central nervous systems (CNSs). Lipid-loaded ApoE lipoprotein particles bind to several cell surface receptors to support membrane homeostasis and brain injury repair. In the brain, ApoE is produced predominantly by astrocytes, but it is also abundantly expressed in most neurons of the CNS. In this study, we addressed the role of ApoE in the hippocampus in mice, focusing on its role in response to radiation injury. To this aim, 8-week-old, wild-type, and ApoE-deficient (ApoE-/-) female mice were acutely whole-body irradiated with 3 Gy of X-rays (0.89 Gy/min), then sacrificed 150 days post-irradiation. In addition, age-matching ApoE-/- females were chronically whole-body irradiated (20 mGy/d, cumulative dose of 3 Gy) for 150 days at the low dose-rate facility at the Institute of Environmental Sciences (IES), Rokkasho, Japan. To seek for ApoE-dependent modification during lineage progression from neural stem cells to neurons, we have evaluated the cellular composition of the dentate gyrus in unexposed and irradiated mice using stage-specific markers of adult neurogenesis. Our findings indicate that ApoE genetic inactivation markedly perturbs adult hippocampal neurogenesis in unexposed and irradiated mice. The effect of ApoE inactivation on the expression of a panel of miRNAs with an established role in hippocampal neurogenesis, as well as its transcriptional consequences in their target genes regulating neurogenic program, have also been analyzed. Our data show that the absence of ApoE-/- also influences synaptic functionality and integration by interfering with the regulation of mir-34a, mir-29b, and mir-128b, leading to the downregulation of synaptic markers PSD95 and synaptophysin mRNA. Finally, compared to acute irradiation, chronic exposure of ApoE null mice yields fewer consequences except for the increased microglia-mediated neuroinflammation. Exploring the function of ApoE in the hippocampus could have implications for developing therapeutic approaches to alleviate radiation-induced brain injury.


Assuntos
Apolipoproteínas E , Hipocampo , MicroRNAs , Radiação Ionizante , Animais , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética , Hipocampo/metabolismo , Hipocampo/efeitos da radiação , Camundongos , Feminino , MicroRNAs/metabolismo , MicroRNAs/genética , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/efeitos da radiação , Neurogênese/efeitos da radiação , Irradiação Corporal Total , Exposição à Radiação/efeitos adversos , Giro Denteado/metabolismo , Giro Denteado/efeitos da radiação , Giro Denteado/patologia
20.
Sci Rep ; 14(1): 12274, 2024 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-38806540

RESUMO

Cranial irradiation used to control brain malignancies invariably leads to progressive and debilitating declines in cognition. Clinical efforts implementing hippocampal avoidance and NMDAR antagonism, have sought to minimize dose to radiosensitive neurogenic regions while normalizing excitatory/inhibitory (E/I) tone. Results of these trials have yielded only marginal benefits to cognition, prompting current studies to evaluate the potential of systemic extracellular vesicle (EV) therapy to restore neurocognitive functionality in the irradiated brain. Here we tested the hypothesis that EVs derived from inhibitory but not excitatory neuronal cultures would prove beneficial to cognition and associated pathology. Rats subjected to a clinically relevant, fractionated cranial irradiation paradigm were given multiple injections of either GABAergic- or glutamatergic-derived EV and subjected to behavioral testing. Rats treated with GABAergic but not glutamatergic EVs showed significant improvements on hippocampal- and cortical-dependent behavioral tasks. While each treatment enhanced levels of the neurotrophic factors BDNF and GDNF, only GABAergic EVs preserved granule cell neuron dendritic spine density. Additional studies conducted with GABAergic EVs, confirmed significant benefits on amygdala-dependent behavior and modest changes in synaptic plasticity as measured by long-term potentiation. These data point to a potentially more efficacious approach for resolving radiation-induced neurological deficits, possibly through a mechanism able to restore homeostatic E/I balance.


Assuntos
Irradiação Craniana , Vesículas Extracelulares , Neurônios GABAérgicos , Animais , Vesículas Extracelulares/metabolismo , Ratos , Irradiação Craniana/efeitos adversos , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/efeitos da radiação , Masculino , Hipocampo/efeitos da radiação , Hipocampo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios/efeitos da radiação , Neurônios/metabolismo , Ácido Glutâmico/metabolismo , Plasticidade Neuronal/efeitos da radiação , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Comportamento Animal/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...