Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
Cell Commun Signal ; 22(1): 216, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38570868

RESUMO

BACKGROUND: Radiation-induced brain injury (RIBI) is a common and severe complication during radiotherapy for head and neck tumor. Repetitive transcranial magnetic stimulation (rTMS) is a novel and non-invasive method of brain stimulation, which has been applied in various neurological diseases. rTMS has been proved to be effective for treatment of RIBI, while its mechanisms have not been well understood. METHODS: RIBI mouse model was established by cranial irradiation, K252a was daily injected intraperitoneally to block BDNF pathway. Immunofluorescence staining, immunohistochemistry and western blotting were performed to examine the microglial pyroptosis and hippocampal neurogenesis. Behavioral tests were used to assess the cognitive function and emotionality of mice. Golgi staining was applied to observe the structure of dendritic spine in hippocampus. RESULTS: rTMS significantly promoted hippocampal neurogenesis and mitigated neuroinflammation, with ameliorating pyroptosis in microglia, as well as downregulation of the protein expression level of NLRP3 inflammasome and key pyroptosis factor Gasdermin D (GSDMD). BDNF signaling pathway might be involved in it. After blocking BDNF pathway by K252a, a specific BDNF pathway inhibitor, the neuroprotective effect of rTMS was markedly reversed. Evaluated by behavioral tests, the cognitive dysfunction and anxiety-like behavior were found aggravated with the comparison of mice in rTMS intervention group. Moreover, the level of hippocampal neurogenesis was found to be attenuated, the pyroptosis of microglia as well as the levels of GSDMD, NLRP3 inflammasome and IL-1ß were upregulated. CONCLUSION: Our study indicated that rTMS notably ameliorated RIBI-induced cognitive disorders, by mitigating pyroptosis in microglia and promoting hippocampal neurogenesis via mediating BDNF pathway.


Assuntos
Lesões Encefálicas , Disfunção Cognitiva , Camundongos , Animais , Estimulação Magnética Transcraniana/efeitos adversos , Estimulação Magnética Transcraniana/métodos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Microglia/metabolismo , Piroptose , Inflamassomos/metabolismo , Encéfalo/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/terapia , Cognição , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Neurogênese/efeitos da radiação
2.
Life Sci Space Res (Amst) ; 35: 170-179, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36336363

RESUMO

Neurogenesis is an essential, lifelong process during which neural stem cells generate new neurons within the hippocampus, a center for learning, memory, and mood control. Neural stem cells are vulnerable to environmental insults spanning from chronic stress to radiation. These insults reduce their numbers and diminish neurogenesis, leading to memory decline, anxiety, and depression. Preserving neural stem cells could thus help prevent these neurogenesis-associated pathologies, an outcome particularly important for long-term space missions where environmental exposure to radiation is significantly higher than on Earth. Multiple developments, from mechanistic discoveries of radiation injury on hippocampal neurogenesis to new platforms for the development of selective, specific, effective, and safe small molecules as neurogenesis-protective agents hold great promise to minimize radiation damage on neurogenesis. In this review, we summarize the effects of space-like radiation on hippocampal neurogenesis. We then focus on current advances in drug discovery and development and discuss the nuclear receptor TLX/NR2E1 (oleic acid receptor) as an example of a neurogenic target that might rescue neurogenesis following radiation.


Assuntos
Astronautas , Lesões por Radiação , Humanos , Neurogênese/fisiologia , Neurogênese/efeitos da radiação , Hipocampo/patologia , Cognição , Lesões por Radiação/prevenção & controle
3.
Int J Mol Sci ; 23(15)2022 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-35955439

RESUMO

Radiation-induced brain injury (RIBI) after radiotherapy has become an increasingly important factor affecting the prognosis of patients with head and neck tumor. With the delivery of high doses of radiation to brain tissue, microglia rapidly transit to a pro-inflammatory phenotype, upregulate phagocytic machinery, and reduce the release of neurotrophic factors. Persistently activated microglia mediate the progression of chronic neuroinflammation, which may inhibit brain neurogenesis leading to the occurrence of neurocognitive disorders at the advanced stage of RIBI. Fully understanding the microglial pathophysiology and cellular and molecular mechanisms after irradiation may facilitate the development of novel therapy by targeting microglia to prevent RIBI and subsequent neurological and neuropsychiatric disorders.


Assuntos
Lesões Encefálicas , Lesões por Radiação , Encéfalo/patologia , Lesões Encefálicas/patologia , Humanos , Microglia/patologia , Neurogênese/efeitos da radiação , Lesões por Radiação/etiologia , Lesões por Radiação/patologia
4.
Sci Rep ; 12(1): 1812, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35110659

RESUMO

The induction and direction of stem cell differentiation into needed cell phenotypes is the central pillar of tissue engineering for repairing damaged tissues or organs. Conventionally, a special recipe of chemical factors is formulated to achieve this purpose for each specific target cell type. In this work, it is demonstrated that the combination of extrinsic photobiomodulation and collagen-covered microislands could be used to induce differentiation of Wharton's jelly mesenchymal stem cells (WJ-MSCs) with the differentiation direction dictated by the specific island topography without use of chemical factors. Both neurogenic differentiation and adipogenic differentiation could be attained with a rate surpassing that using chemical factors. Application of this method to other cell types is possible by utilizing microislands with a pattern tailored particularly for each specific cell type, rendering it a versatile modality for initiating and guiding stem cell differentiation.


Assuntos
Adesão Celular , Diferenciação Celular/efeitos da radiação , Colágeno/fisiologia , Luz , Células-Tronco Mesenquimais/efeitos da radiação , Engenharia Tecidual , Adipogenia/efeitos da radiação , Técnicas de Cultura de Células , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/fisiologia , Neurogênese/efeitos da radiação , Geleia de Wharton/citologia
5.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34830484

RESUMO

Radiation therapy represents one of the primary treatment modalities for primary and metastatic brain tumors. Although recent advances in radiation techniques, that allow the delivery of higher radiation doses to the target volume, reduce the toxicity to normal tissues, long-term neurocognitive decline is still a detrimental factor significantly affecting quality of life, particularly in pediatric patients. This imposes the need for the development of prevention strategies. Based on recent evidence, showing that manipulation of the Shh pathway carries therapeutic potential for brain repair and functional recovery after injury, here we evaluate how radiation-induced hippocampal alterations are modulated by the constitutive activation of the Shh signaling pathway in Patched 1 heterozygous mice (Ptch1+/-). Our results show, for the first time, an overall protective effect of constitutive Shh pathway activation on hippocampal radiation injury. This activation, through modulation of the proneural gene network, leads to a long-term reduction of hippocampal deficits in the stem cell and new neuron compartments and to the mitigation of radio-induced astrogliosis, despite some behavioral alterations still being detected in Ptch1+/- mice. A better understanding of the pathogenic mechanisms responsible for the neural decline following irradiation is essential for identifying prevention measures to contain the harmful consequences of irradiation. Our data have important translational implications as they suggest a role for Shh pathway manipulation to provide the therapeutic possibility of improving brain repair and functional recovery after radio-induced injury.


Assuntos
Proteínas Hedgehog/genética , Hipocampo/efeitos da radiação , Neurogênese/genética , Receptor Patched-1/genética , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Redes Reguladoras de Genes/efeitos da radiação , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Camundongos , Camundongos Knockout , Neurogênese/efeitos da radiação , Neurônios/metabolismo , Neurônios/efeitos da radiação , Qualidade de Vida , Radiação Ionizante , Transdução de Sinais/efeitos da radiação
6.
Int J Radiat Biol ; 97(7): 926-942, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34043460

RESUMO

ABSTRACТPurpose: To explore if the total body γ-irradiation at a dose of 0.1 Gy 7 days prior to acute mixed γ, n-irradiation of the head at the dose of 1 Gy can reduce the harmful effects of neutron irradiation on the hippocampal functions, neuroinflammation and neurogenesis.Materials and methods: Mice were exposed to γ-radiation alone, mixed γ,n-radiation or combined γ-rays and γ,n-radiation 7 days after γ-irradiation. Two months post-irradiation, mice were tested in Open Field and in the Morris water maze. The content of microglia, astrocytes, proliferating cells and cytokines TGF-ß, TNF-α, IL-1ß, GFAP levels, hippocampal BDNF, NT-3, NT-4, NGF mRNA expression were evaluated.Results: Two months after combined irradiation, we observed impaired hippocampus-dependent cognition, which was not detected in mice exposed to γ,n-irradiation. Combined exposure and γ,n-irradiation led to a significant increase in the level of activated microglia and astrocytes in the brains. The level of pro- and anti-inflammatory cytokines in the brain and hippocampal neurotrophine's genes changed differenly after the combined exposure and γ,n-irradiation. The quantity of DCX-positive cells was reduced after γ,n-irradiation exposer alone, but increased after combined irradiation.Conclusions: Our results indicate radio-adaptive responses in brains of mice that were exposed to low-dose gamma irradiation 7 days prior to acute 1 Gy γ,n-irradiation.


Assuntos
Sistema Nervoso Central/efeitos da radiação , Raios gama , Nêutrons , Animais , Sistema Nervoso Central/metabolismo , Citocinas/metabolismo , Relação Dose-Resposta à Radiação , Proteína Duplacortina , Cabeça/efeitos da radiação , Camundongos , Neurogênese/efeitos da radiação
7.
BMC Neurosci ; 22(1): 28, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33882822

RESUMO

BACKGROUND: Brain radiation exposure, in particular, radiotherapy, can induce cognitive impairment in patients, with significant effects persisting for the rest of their life. However, the main mechanisms leading to this adverse event remain largely unknown. A study of radiation-induced injury to multiple brain regions, focused on the hippocampus, may shed light on neuroanatomic bases of neurocognitive impairments in patients. Hence, we irradiated BALB/c mice (male and female) at postnatal day 3 (P3), day 10 (P10), and day 21 (P21) and investigated the long-term radiation effect on brain MRI changes and hippocampal neurogenesis. RESULTS: We found characteristic brain volume reductions in the hippocampus, olfactory bulbs, the cerebellar hemisphere, cerebellar white matter (WM) and cerebellar vermis WM, cingulate, occipital and frontal cortices, cerebellar flocculonodular WM, parietal region, endopiriform claustrum, and entorhinal cortex after irradiation with 5 Gy at P3. Irradiation at P10 induced significant volume reduction in the cerebellum, parietal region, cingulate region, and olfactory bulbs, whereas the reduction of the volume in the entorhinal, parietal, insular, and frontal cortices was demonstrated after irradiation at P21. Immunohistochemical study with cell division marker Ki67 and immature marker doublecortin (DCX) indicated the reduced cell division and genesis of new neurons in the subgranular zone of the dentate gyrus in the hippocampus after irradiation at all three postnatal days, but the reduction of total granule cells in the stratum granulosun was found after irradiation at P3 and P10. CONCLUSIONS: The early life radiation exposure during different developmental stages induces varied brain pathophysiological changes which may be related to the development of neurological and neuropsychological disorders later in life.


Assuntos
Encéfalo/efeitos da radiação , Irradiação Craniana/efeitos adversos , Neurogênese/efeitos da radiação , Animais , Animais Recém-Nascidos , Encéfalo/crescimento & desenvolvimento , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C
8.
Int J Mol Sci ; 22(8)2021 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-33924260

RESUMO

The brain undergoes ionizing radiation exposure in many clinical situations, particularly during radiotherapy for brain tumors. The critical role of the hippocampus in the pathogenesis of radiation-induced neurocognitive dysfunction is well recognized. The goal of this study is to test the potential contribution of non-targeted effects in the detrimental response of the hippocampus to irradiation and to elucidate the mechanisms involved. C57Bl/6 mice were whole body (WBI) or partial body (PBI) irradiated with 0.1 or 2.0 Gy of X-rays or sham irradiated. PBI consisted of the exposure of the lower third of the mouse body, whilst the upper two thirds were shielded. Hippocampi were collected 15 days or 6 months post-irradiation and a multi-omics approach was adopted to assess the molecular changes in non-coding RNAs, proteins and metabolic levels, as well as histological changes in the rate of hippocampal neurogenesis. Notably, at 2.0 Gy the pattern of early molecular and histopathological changes induced in the hippocampus at 15 days following PBI were similar in quality and quantity to the effects induced by WBI, thus providing a proof of principle of the existence of out-of-target radiation response in the hippocampus of conventional mice. We detected major alterations in DAG/IP3 and TGF-ß signaling pathways as well as in the expression of proteins involved in the regulation of long-term neuronal synaptic plasticity and synapse organization, coupled with defects in neural stem cells self-renewal in the hippocampal dentate gyrus. However, compared to the persistence of the WBI effects, most of the PBI effects were only transient and tended to decrease at 6 months post-irradiation, indicating important mechanistic difference. On the contrary, at low dose we identified a progressive accumulation of molecular defects that tended to manifest at later post-irradiation times. These data, indicating that both targeted and non-targeted radiation effects might contribute to the pathogenesis of hippocampal radiation-damage, have general implications for human health.


Assuntos
Irradiação Craniana , Hipocampo/metabolismo , Hipocampo/efeitos da radiação , Metaboloma , Neurogênese/genética , Neurogênese/efeitos da radiação , Proteoma , Transcriptoma , Animais , Biologia Computacional/métodos , Irradiação Craniana/efeitos adversos , Feminino , Regulação da Expressão Gênica , Imuno-Histoquímica , Camundongos , Doses de Radiação , Transdução de Sinais
9.
Sci Rep ; 11(1): 4292, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33619310

RESUMO

Galactic cosmic radiation (GCR) composed of high-energy, heavy particles (HZE) poses potentially serious hazards to long-duration crewed missions in deep space beyond earth's magnetosphere, including planned missions to Mars. Chronic effects of GCR exposure on brain structure and cognitive function are poorly understood, thereby limiting risk reduction and mitigation strategies to protect against sequelae from exposure during and after deep-space travel. Given the selective vulnerability of the hippocampus to neurotoxic insult and the importance of this brain region to learning and memory, we hypothesized that GCR-relevant HZE exposure may induce long-term alterations in adult hippocampal neurogenesis, synaptic plasticity, and hippocampal-dependent learning and memory. To test this hypothesis, we irradiated 3-month-old male and female mice with a single, whole-body dose of 10, 50, or 100 cGy 56Fe ions (600 MeV, 181 keV/µm) at Brookhaven National Laboratory. Our data reveal complex, dynamic, time-dependent effects of HZE exposure on the hippocampus. Two months post exposure, neurogenesis, synaptic plasticity and learning were impaired compared to sham-irradiated, age-matched controls. By six months post-exposure, deficits in spatial learning were absent in irradiated mice, and synaptic potentiation was enhanced. Enhanced performance in spatial learning and facilitation of synaptic plasticity in irradiated mice persisted 12 months post-exposure, concomitant with a dramatic rebound in adult-born neurons. Synaptic plasticity and spatial learning remained enhanced 20 months post-exposure, indicating a life-long influence on plasticity and cognition from a single exposure to HZE in young adulthood. These findings suggest that GCR-exposure can persistently alter brain health and cognitive function during and after long-duration travel in deep space.


Assuntos
Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Radiação Cósmica/efeitos adversos , Astronautas , Biomarcadores , Encéfalo/fisiopatologia , Giro Denteado/metabolismo , Giro Denteado/fisiopatologia , Giro Denteado/efeitos da radiação , Exposição Ambiental/efeitos adversos , Feminino , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Hipocampo/efeitos da radiação , Humanos , Masculino , Neurogênese/efeitos da radiação , Exposição à Radiação/efeitos adversos , Voo Espacial , Aprendizagem Espacial/efeitos da radiação , Fatores de Tempo
10.
Nat Commun ; 12(1): 1174, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33608552

RESUMO

Adult hippocampal neurogenesis contributes to learning and memory, and is sensitive to a variety of environmental stimuli. Exposure to a hypomagnetic field (HMF) influences the cognitive processes of various animals, from insects to human beings. However, whether HMF exposure affect adult hippocampal neurogenesis and hippocampus-dependent cognitions is still an enigma. Here, we showed that male C57BL/6 J mice exposed to HMF by means of near elimination of the geomagnetic field (GMF) exhibit significant impairments of adult hippocampal neurogenesis and hippocampus-dependent learning, which is strongly correlated with a reduction in the content of reactive oxygen species (ROS). However, these deficits seen in HMF-exposed mice could be rescued either by elevating ROS levels through pharmacological inhibition of ROS removal or by returning them back to GMF. Therefore, our results suggest that GMF plays an important role in adult hippocampal neurogenesis through maintaining appropriate endogenous ROS levels.


Assuntos
Cognição/fisiologia , Cognição/efeitos da radiação , Hipocampo/patologia , Hipocampo/efeitos da radiação , Campos Magnéticos/efeitos adversos , Neurogênese/fisiologia , Neurogênese/efeitos da radiação , Adulto , Animais , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Aprendizagem , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais , Neurogênese/genética , Espécies Reativas de Oxigênio
11.
Int J Radiat Biol ; 97(3): 329-340, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33332177

RESUMO

PURPOSE: Despite being a major treatment modality for brain cancer due to its efficiency in achieving cancer control, radiotherapy has long been known to cause long-term side effects, including radiation-induced cognitive impairment (RICI). Neurogenesis inhibition due to radiation-induced damage in neural stem cells (NSCs) has been demonstrated to be an important mechanism underlying RICI. Radiation-induced bystander effects (RIBEs) denote the biological responses in non-targeted cells after their neighboring cells are irradiated. We have previously demonstrated that RIBEs could play an important role in the skin wound healing process. Therefore, we aimed to investigate whether RIBEs contribute to RICI in this study. MATERIALS AND METHODS: The transwell co-culture method was used to investigate bystander effects in mouse NSCs induced by irradiated GL261 mouse glioma cells in vitro. The proliferation, neurosphere-forming capacity and differentiation potential of NSCs were determined as the bystander endpoints. The exosomes were extracted from the media used to culture GL261 cells and were injected into the hippocampus of C57BL/6 mice. Two months later, the neurogenesis of mice was assessed using BrdU incorporation and immunofluorescence microscopy, and cognitive function was evaluated by the Morris Water Maze. RESULTS: After co-culture with GL261 glioma cells, mouse NSCs displayed inhibited proliferation and reduced neurosphere-forming capacity and differentiation potential. The irradiated GL261 cells caused greater inhibition and reduction in NSCs than unirradiated GL261 cells. Moreover, adding the exosomes secreted by GL261 cells into the culture of NSCs inhibited NSC proliferation, suggesting that the cancer cell-derived exosomes may be critical intercellular signals. Furthermore, injection of the exosomes from GL261 cells into the hippocampus of mice caused significant neurogenesis inhibition and cognitive impairment two month later, and the exosomes from irradiated GL261 cells induced greater inhibitory effects. CONCLUSION: RIBEs mediated by the exosomes from irradiated cancer cells could contribute to RICI and, therefore, could be a novel mechanism underlying RICI.


Assuntos
Neoplasias Encefálicas/radioterapia , Efeito Espectador/efeitos da radiação , Disfunção Cognitiva/etiologia , Glioma/radioterapia , Células-Tronco Neurais/efeitos da radiação , Animais , Neoplasias Encefálicas/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Exossomos/fisiologia , Feminino , Glioma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese/efeitos da radiação
12.
Oncol Rep ; 44(6): 2527-2536, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33125501

RESUMO

Cognitive deficit is one of the most serious complications of cranial radiotherapy of head and neck cancers. However, the underlying mechanism of this cognitive impairment remains unclear. In the present study, the role of tropomyosin receptor kinase A (TrkA) and its ligand neurotrophin nerve growth factor (NGF) were investigated following whole­brain irradiation (WBI). Young male Sprague­Dawley rats underwent WBI at a single dose of 10 Gy. WBI was determined to result in notable memory decline and substantial neurogenesis impairment in the hippocampus 3 months post­irradiation. Compared with the control group, TrkA protein expression was greater in irradiated rats 1 week after WBI, which then decreased significantly by the 3­month time­point. However, no difference in NGF expression was observed from 1 day to 3 months post­WBI. Overexpression of hippocampal TrkA in rats using adeno­associated virus ameliorated memory decline induced by irradiation. Additionally, upregulating TrkA expression rescued irradiation­induced hippocampal precursor cell proliferation and promoted neurogenesis. PI3K, Akt and ERK1/2 phosphorylation were also revealed to be significantly inhibited by WBI, which was ameliorated by TrkA overexpression. Findings of the present study indicated that the TrkA­dependent signaling pathway may serve a critical role in radiotherapy­induced cognitive deficit and impairments in neurogenesis.


Assuntos
Disfunção Cognitiva/patologia , Irradiação Craniana/efeitos adversos , Hipocampo/patologia , Lesões Experimentais por Radiação/patologia , Receptor trkA/metabolismo , Animais , Disfunção Cognitiva/etiologia , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Neoplasias de Cabeça e Pescoço/radioterapia , Hipocampo/efeitos da radiação , Humanos , Masculino , Fator de Crescimento Neural/metabolismo , Neurogênese/genética , Neurogênese/efeitos da radiação , Lesões Experimentais por Radiação/etiologia , Ratos , Receptor trkA/genética , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Técnicas Estereotáxicas
13.
Cells ; 9(9)2020 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-32933226

RESUMO

It is a well-known fact that following a proper routine light/dark or diurnal rhythm controls almost all biological processes. With the introduction of modern lighting and artificial illumination systems, continuous exposure to light at night may lead to the disruption of diurnal rhythm. However, the effect of light during the night on brain anatomy, physiology, and human body functions is less explored and poorly understood. In this study, we have evaluated the effect of exposure to dim light (5 lux) at night (dLAN) on Swiss Albino mice over a duration of three consecutive weeks. Results have revealed that exposure to dLAN led to an impairment of cognitive and non-cognitive behaviour, oxidative stress-mediated elevation of lipid peroxidation, and reduction of superoxide dismutase and catalase activity. It also led to the downregulation of hippocampal proteins (BDNF, Synapsin II and DCX) at both protein and mRNA level. Additionally, there was downregulation of CREB and SIRT1 mRNAs and neurodegeneration-associated miRNA21a-5p and miRNA34a-5p. The pyramidal and cortical neurons started showing pyknotic and chromatolysis characteristics. However, a dose of curcumin administered to the mice positively modulated these parameters in our experimental animals. We proposed the modulatory role of curcumin in addressing the deleterious effects of dLAN.


Assuntos
Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/efeitos da radiação , Curcumina/farmacologia , Luz/efeitos adversos , Fármacos Neuroprotetores/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/efeitos da radiação , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Cognição/efeitos da radiação , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Hipocampo/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurogênese/efeitos dos fármacos , Neurogênese/efeitos da radiação , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Sinapsinas/genética , Sinapsinas/metabolismo
14.
Brain Res ; 1748: 147095, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32896524

RESUMO

Cranial and craniospinal irradiation are the oldest central nervous system prophylaxis treatments considered for pediatric patients with acute lymphoblastic leukemia (ALL). However, survivors of childhood ALL that received cranial radiotherapy are at increased risk for deficits in neurocognitive skills. The continuous and dynamic response of normal tissue after irradiation has been identified as one of the causative factors for cognitive changes after cranial radiation therapy. The aim of our study was to investigate the radiation effects on social behavior and neuronal morphology in the hippocampus of adult mice. Twenty-oneday-old male C57BL/6 mice were irradiated with the small-animal radiation research platform (SARRP). Animals were given a single 10-Gy dose of radiation of X-ray cranial radiation. One month following irradiation, animals underwent behavioral testing in the Three-Chamber Sociability paradigm. Radiation affected social discrimination during the third stage eliciting an inability to discriminate between the familiar and stranger mouse, while sham successfully spent more time exploring the novel stranger. Proteomic analysis revealed dysregulation of metabolic and signaling pathways associated with neurocognitive dysfunction such as mitochondrial dysfunction, Rac 1 signaling, and synaptogenesis signaling. We observed significant decreases in mushroom spine density in the Cornu Ammonis 2 of the hippocampus, which is associated with sociability processing.


Assuntos
Comportamento Animal/efeitos da radiação , Irradiação Craniana , Hipocampo/efeitos da radiação , Memória/efeitos da radiação , Comportamento Social , Animais , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/efeitos da radiação , Hipocampo/metabolismo , Masculino , Camundongos , Neurogênese/efeitos da radiação , Proteômica , Transdução de Sinais/efeitos da radiação , Sirtuínas/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
15.
Neurotoxicology ; 79: 40-47, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32320710

RESUMO

Ionizing radiation (IR) is increasingly used for diagnostics and therapy of severe brain diseases. However, IR also has adverse effects on the healthy brain tissue, particularly on the neuronal network. This is true for adults but even more pronounced in the developing brain of unborn and pediatric patients. Epidemiological studies on children receiving radiotherapy showed an increased risk for cognitive decline ranging from mild deficits in academic functioning to severe late effects in intellectual ability and language as a consequence of altered neuronal development and connectivity. To provide a comprehensive approach for the analysis of radiation-induced alterations in human neuronal functionality, we developed an in vitro assay by combining microelectrode array (MEA) analyses and human embryonic stem cell (hESC) derived three-dimensional neurospheres (NS). In our proof of principle study, we irradiated hESC with 1 Gy X-rays and let them spontaneously differentiate into neurons within NS. After the onset of neuronal activity, we recorded and analyzed the activity pattern of the developing neuronal networks. The network activity in NS derived from irradiated hESC was significantly reduced, whereas no differences in molecular endpoints such as cell proliferation and transcript or protein expression analyses were found. Thus, the combination of MEA analysis with a 3D model for neuronal functionality revealed radiation sequela that otherwise would not have been detected. We therefore strongly suggest combining traditional biomolecular methods with the new functional assay presented in this work to improve the risk assessment for IR-induced effects on the developing brain.


Assuntos
Células-Tronco Embrionárias Humanas/efeitos da radiação , Rede Nervosa/efeitos da radiação , Células-Tronco Neurais/efeitos da radiação , Neurogênese/efeitos da radiação , Potenciais de Ação/efeitos dos fármacos , Técnicas de Cultura de Células/instrumentação , Proliferação de Células/efeitos da radiação , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/instrumentação , Rede Nervosa/metabolismo , Células-Tronco Neurais/metabolismo , Fenótipo , Estudo de Prova de Conceito , Esferoides Celulares
16.
Nanoscale ; 12(17): 9833-9841, 2020 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-32342083

RESUMO

Nerve tissues are one of the most difficult tissues to repair due to the limited source of neural stem cells and the difficulty in promoting the neural differentiation of mesenchymal stem cells by growth factors. Electromagnetic field has been proved to have the ability to regulate stem cell differentiation. Although some research studies promoted the neural differentiation of stem cells using an external power source, it is still a big challenge to realize nerve repair in bodies because of the unwieldiness and complexity of the power supply equipment. Surface plasmons (SP) are electromagnetic oscillations caused by the interaction of free electrons and photons on a metal surface, and almost no one has used these localized electromagnetic oscillations to regulate stem cell differentiation. In this study, based on the concept proposed by our group that "the stem cell fate can be regulated by nanostructure mediated physical signals", the localized electromagnetic oscillation generated by the localized surface plasmon resonance (LSPR) of copper sulfide (CuS) nanostructures irradiated with near-infrared light has been proved to have positive regulation on stem cell maturation and neuron-like cell differentiation of human adipose-derived stem cells (hADSCs). This regulation method avoids the use of wire connection of an external power source, which realizes the stem cell fate regulation by an external field. In addition, this work demonstrated that it is promising to realize the light promoted nerve repair in bodies by using an implantable plasmonic nanomaterial with absorption in the near-infrared region within a human "optical window", which has important academic value and application prospect. As we know, this is the first time to use semiconductor nanostructures as a medium to regulate stem cell neuron-like cell differentiation by near-infrared light and the LSPR of a plasmonic nanomaterial, which will have great influence on biomedical engineering and attract broad attention from nanomaterials scientists, neurobiologists, and neurosurgeons.


Assuntos
Cobre/química , Nanoestruturas/química , Neurogênese/efeitos da radiação , Células-Tronco/citologia , Tecido Adiposo/citologia , Adesão Celular , Sobrevivência Celular , Células Cultivadas , Cobre/metabolismo , Cobre/efeitos da radiação , Humanos , Luz , Nanoestruturas/efeitos da radiação , Células-Tronco/efeitos da radiação , Ressonância de Plasmônio de Superfície
17.
Radiat Res ; 193(5): 460-470, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32216708

RESUMO

Human embryonic brain development is highly sensitive to ionizing radiation. However, detailed information on the mechanisms of this sensitivity is not available due to limited experimental data. In this study, differentiation of human embryonic stem cells (hESCs) to neural lineages was used as a model for early embryonic brain development to assess the effect of exposure to low (17 mGy) and high (572 mGy) doses of radiation on gene expression. Transcriptomes were assessed using RNA sequencing during neural differentiation at three time points in control and irradiated samples. The first time point was when the cells were still pluripotent (day 0), the second time point was during the stage of embryoid body formation (day 6), and the third and final time point was during the stage of neural rosette formation (day 10). Analysis of the transcriptomes revealed neurodifferentiation in both the control and irradiated cells. Low-dose irradiation did not result in changes in gene expression at any of the time points, whereas high-dose irradiation resulted in downregulation of some major neurodifferentiation markers on days 6 and 10. Gene ontology analysis showed that pathways related to nervous system development, neurogenesis and generation of neurons were among the most affected. Expression of such key regulators of neuronal development as NEUROG1, ARX, ASCL1, RFX4 and INSM1 was reduced more than twofold. In conclusion, exposure to a 17 mGy low dose of radiation was well tolerated by hESCs while exposure to 572 mGy significantly affected their genetic reprogramming into neuronal lineages.


Assuntos
Diferenciação Celular/efeitos da radiação , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/efeitos da radiação , Transcriptoma/efeitos da radiação , Células-Tronco Embrionárias Humanas/citologia , Humanos , Neurogênese/efeitos da radiação , Fatores de Tempo , Tomografia Computadorizada por Raios X/efeitos adversos
18.
Gene ; 738: 144485, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32087272

RESUMO

Impairment of neurogenesis is thought to be one of the important mechanisms underlying radiation-induced cognitive decline. Self-renewal and differentiation of neural stem cells (NSCs) are important components of neurogenesis. It has been well established that autophagy plays an important role in neurodegenerative conditions, however, its role in radiation-induced cognitive decline remains unclear. Our previous studies have found that ionizing radiation (IR) induces autophagy in mouse neurons, and minocycline, an antibiotic that can cross the blood-brain barrier, protects neurons from radiation-induced apoptosis through promoting autophagy, thus may contribute to the improvement of mouse cognitive performance after whole-brain irradiation. In the present study, we investigated whether autophagy is involved in radiation-induced damage in self-renewal and differentiation of NSCs. We found that NSCs were extremely sensitive to IR. Irradiation induced autophagy in NSCs in a dose-dependent manner. Atg7 knockdown significantly decreased autophagy, thus increased the apoptosis levels in irradiated NSCs, suggesting that autophagy protected NSCs from radiation-induced apoptosis. Moreover, compared with the negative control NSCs, the neurosphere size was significantly reduced and the neuronal differentiation was notably inhibited in Atg7-deficient NSCs after irradiation, indicating that autophagy defect could exacerbate radiation-induced reduction in NSC self-renewal and differentiation potential. In conclusion, down-regulating autophagy by selective Atg7 knockdown in NSCs enhanced radiation-induced NSC damage, suggesting an important protective role of autophagy in maintaining neurogenesis. Along with the protective effect of autophagy on irradiated neurons, our results on NSCs not only shed the light on the involvement of autophagy in the development of radiation-induced cognitive decline, but also provided a potential target for preventing cognitive impairment after cranial radiation exposure.


Assuntos
Proteína 7 Relacionada à Autofagia/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Autofagia/efeitos da radiação , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Neurogênese/efeitos da radiação , Neurônios/efeitos da radiação , Gravidez , Radiação Ionizante
19.
Epigenomics ; 12(5): 385-396, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32041423

RESUMO

Aim: Roles of forced running exercise (FE) in remediation of neurogenesis inhibition and radiation-induced cognitive dysfunction were investigated in a whole-brain irradiation mice model via the regulation of DNA 5-hydroxymethylation modification (5 hmC) and its catalytic enzymes ten-eleven translocation (Tet) proteins. Materials & methods: Hippocampal neurogenesis and cognitive function, DNA 5 hmC level and Tet expression were determined in mice. Results: The expression of DNA 5 hmC and Tet2, brain-derived neurotrophic factor significantly decreased in hippocampus postradiation. FE mitigated radiation-induced neurogenesis deficits and cognitive dysfunction. Furthermore, FE increased 5 hmC and brain-derived neurotrophic factor expression. SC1, a Tet inhibitor, reversed partly such changes. Conclusion: Tet-mediated 5 hmC modification represents a kind of diagnostic biomarkers of radiation-induced cognitive dysfunction. Targeting Tet-related epigenetic modification may be a novel therapeutic strategy for radiation-induced brain injury.


Assuntos
Cognição , Disfunção Cognitiva/etiologia , Metilação de DNA/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Condicionamento Físico Animal/efeitos adversos , Radiação Ionizante , 5-Metilcitosina , Animais , Biomarcadores , Disfunção Cognitiva/diagnóstico , Irradiação Craniana , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Epigênese Genética , Hipocampo/metabolismo , Camundongos , Neurogênese/genética , Neurogênese/efeitos da radiação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Lesões por Radiação/diagnóstico , Lesões por Radiação/etiologia
20.
Neuromolecular Med ; 22(1): 139-149, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31595404

RESUMO

Optogenetic stimulation of neural stem cells (NSCs) enables their activity-dependent photo-modulation. This provides a spatio-temporal tool for studying activity-dependent neurogenesis and for regulating the differentiation of the transplanted NSCs. Currently, this is mainly driven by viral transfection of channelrhodopsin-2 (ChR2) gene, which requires high irradiance and complex in vivo/vitro stimulation systems. Additionally, despite the extensive application of optogenetics in neuroscience, the transcriptome-level changes induced by optogenetic stimulation of NSCs have not been elucidated yet. Here, we made transformed NSCs (SFO-NSCs) stably expressing one of the step-function opsin (SFO)-variants of chimeric channelrhodopsins, ChRFR(C167A), which is more sensitive to blue light than native ChR2, via a non-viral transfection system using piggyBac transposon. We set up a simple low-irradiance optical stimulation (OS)-incubation system that induced c-fos mRNA expression, which is activity-dependent, in differentiating SFO-NSCs. More neuron-like SFO-NCSs, which had more elongated axons, were differentiated with daily OS than control cells without OS. This was accompanied by positive/negative changes in the transcriptome involved in axonal remodeling, synaptic plasticity, and microenvironment modulation with the up-regulation of several genes involved in the Ca2+-related functions. Our approach could be applied for stem cell transplantation studies in tissue with two strengths: lower carcinogenicity and less irradiance needed for tissue penetration.


Assuntos
Células-Tronco Neurais/efeitos da radiação , Neurogênese/efeitos da radiação , Optogenética , Sinalização do Cálcio , Linhagem Celular Transformada , Channelrhodopsins/biossíntese , Channelrhodopsins/genética , Channelrhodopsins/efeitos da radiação , Elementos de DNA Transponíveis , Regulação da Expressão Gênica/efeitos da radiação , Ontologia Genética , Genes Reporter , Genes fos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/citologia , Plasticidade Neuronal/efeitos da radiação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transcriptoma/efeitos da radiação , Regulação para Cima/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA