Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.291
Filtrar
Más filtros











Intervalo de año de publicación
1.
Food Res Int ; 188: 114433, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38823827

RESUMEN

Whey derived peptides have shown potential activity improving brain function in pathological condition. However, there is little information about their mechanism of action on glial cells, which have important immune functions in brain. Astrocytes and microglia are essential in inflammatory and oxidative defense that take place in neurodegenerative disease. In this work we evaluate antioxidant and anti-inflammatory potential bioactivity of whey peptide in glial cells. Peptides were formed during simulated gastrointestinal digestion (Infogest protocol), and low molecular weight (<5kDA) peptides (WPHf) attenuated reactive oxygen species (ROS) production induced by hydrogen peroxide stimulus in both cells in dose-dependent manner. WPHf induced an increase in the antioxidant glutathione (GSH) content and prevented GSH reduction induced by lipopolysaccharides (LPS) stimulus in astrocytes cells in a cell specific form. An increase in cytokine mRNA expression (TNFα and IL6) and nitric oxide secretion induced by LPS was attenuated by WPHf pre-treatment in both cells. The inflammatory pathway was dependent on NFκB activation. Bioactive peptide ranking analysis showed positive correlation with hydrophobicity and negative correlation with high molecular weights. The sequence identification revealed 19 peptides cross-referred with bioactive database. Whey peptides were rich in leucine, valine and tyrosine in the C-terminal region and lysine in the N-terminal region. The anti-inflammatory and antioxidant potential of whey peptides were assessed in glia cells and its mechanisms of action were related, such as modulation of antioxidant enzymes and anti-inflammatory pathways. Features of the peptide structure, such as molecular size, hydrophobicity and types of amino acids present in the terminal region are associated to bioactivity.


Asunto(s)
Antiinflamatorios , Antioxidantes , Neuroglía , Proteína de Suero de Leche , Antioxidantes/farmacología , Antiinflamatorios/farmacología , Proteína de Suero de Leche/farmacología , Proteína de Suero de Leche/química , Proteína de Suero de Leche/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Animales , Especies Reactivas de Oxígeno/metabolismo , Lipopolisacáridos/farmacología , Glutatión/metabolismo , Péptidos/farmacología , Óxido Nítrico/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo
2.
PeerJ ; 12: e17541, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38832034

RESUMEN

Introduction: Oxidative and antioxidant pathways play essential roles in the development of alcohol-induced brain injury. The Nrf2 pathway is an endogenous antioxidant response pathway, but there has been little research on the role of Nrf2 in alcohol-related diseases. Thus, we examined the effects of alcohol and an Nrf2 agonist (TBHQ) on astrocyte function, mRNA expression, and metabolite content to further explore the protective mechanisms of Nrf2 agonists in astrocytes following alcohol exposure. Methods: CTX TNA2 astrocytes were cultured with alcohol and TBHQ and then subjected to transcriptome sequencing, LC-MS/MS analysis, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and malondialdehyde (MDA) and superoxide dismutase (SOD) activity assays. Results: Alcohol exposure significantly increased malondialdehyde (MDA) levels while decreasing superoxide dismutase (SOD) levels in astrocytes. Treatment with TBHQ effectively reversed these effects, demonstrating its protective role against oxidative stress induced by alcohol. Transcriptome sequencing and qRT-PCR analysis revealed that TBHQ specifically upregulates genes involved in glutathione metabolism, including a notable increase in the expression of the glutathione S-transferase A5 (GSTA5) gene, which was suppressed by alcohol exposure. Additionally, metabolomic analysis showed that TBHQ regulates key components of ether lipid metabolism in alcohol-exposed astrocytes, with significant reductions in the levels of lysophosphatidylcholine (18:0) (LysoPC (18:0)) and 2-acetyl-1-alkyl-sn-glycero-3-phosphocholine, both of which are critical markers in the ether lipid metabolic pathway. Discussion: The findings underscore the role of TBHQ as an Nrf2 agonist in mitigating alcohol-induced oxidative damage in astrocytes by modulating glutathione metabolism and ether lipid metabolism. The regulation of GSTA5 gene expression emerges as a key mechanism through which Nrf2 agonists confer neuroprotection against oxidative stress and lipid oxidation. These insights pave the way for potential therapeutic strategies targeting the Nrf2 pathway to protect astrocytes from alcohol-induced damage.


Asunto(s)
Astrocitos , Etanol , Glutatión , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Animales , Etanol/farmacología , Estrés Oxidativo/efectos de los fármacos , Glutatión/metabolismo , Hidroquinonas/farmacología , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/genética , Malondialdehído/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Células Cultivadas
3.
Sci Rep ; 14(1): 10433, 2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714696

RESUMEN

Toxoplasma gondii (T. gondii) is a protozoan parasite that infects approximately one-third of the global human population, often leading to chronic infection. While acute T. gondii infection can cause neural damage in the central nervous system and result in toxoplasmic encephalitis, the consequences of T. gondii chronic infection (TCI) are generally asymptomatic. However, emerging evidence suggests that TCI may be linked to behavioral changes or mental disorders in hosts. Astrocyte polarization, particularly the A1 subtype associated with neuronal apoptosis, has been identified in various neurodegenerative diseases. Nevertheless, the role of astrocyte polarization in TCI still needs to be better understood. This study aimed to establish a mouse model of chronic TCI and examine the transcription and expression levels of glial fibrillary acidic protein (GFAP), C3, C1q, IL-1α, and TNF-α in the brain tissues of the mice. Quantitative real-time PCR (qRT-PCR), enzyme-linked immunosorbent assay, and Western blotting were employed to assess these levels. Additionally, the expression level of the A1 astrocyte-specific marker C3 was evaluated using indirect fluorescent assay (IFA). In mice with TCI, the transcriptional and expression levels of the inflammatory factors C1q, IL-1α, and TNF-α followed an up-down-up pattern, although they remained elevated compared to the control group. These findings suggest a potential association between astrocyte polarization towards the A1 subtype and synchronized changes in these three inflammatory mediators. Furthermore, immunofluorescence assay (IFA) revealed a significant increase in the A1 astrocytes (GFAP+C3+) proportion in TCI mice. This study provides evidence that TCI can induce astrocyte polarization, a biological process that may be influenced by changes in the levels of three inflammatory factors: C1q, IL-1α, and TNF-α. Additionally, the release of neurotoxic substances by A1 astrocytes may be associated with the development of TCI.


Asunto(s)
Astrocitos , Encéfalo , Toxoplasma , Animales , Astrocitos/metabolismo , Astrocitos/parasitología , Astrocitos/patología , Ratones , Toxoplasma/patogenicidad , Toxoplasma/fisiología , Encéfalo/parasitología , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Enfermedad Crónica , Polaridad Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Toxoplasmosis/metabolismo , Toxoplasmosis/parasitología , Toxoplasmosis/patología , Factor de Necrosis Tumoral alfa/metabolismo , Toxoplasmosis Cerebral/parasitología , Toxoplasmosis Cerebral/patología , Toxoplasmosis Cerebral/metabolismo
4.
Int J Neural Syst ; 34(6): 2450028, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38706265

RESUMEN

Spiking neural membrane systems (or spiking neural P systems, SNP systems) are a new type of computation model which have attracted the attention of plentiful scholars for parallelism, time encoding, interpretability and extensibility. The original SNP systems only consider the time delay caused by the execution of rules within neurons, but not caused by the transmission of spikes via synapses between neurons and its adaptive adjustment. In view of the importance of time delay for SNP systems, which are a time encoding computation model, this study proposes SNP systems with adaptive synaptic time delay (ADSNP systems) based on the dynamic regulation mechanism of synaptic transmission delay in neural systems. In ADSNP systems, besides neurons, astrocytes that can generate adenosine triphosphate (ATP) are introduced. After receiving spikes, astrocytes convert spikes into ATP and send ATP to the synapses controlled by them to change the synaptic time delays. The Turing universality of ADSNP systems in number generating and accepting modes is proved. In addition, a small universal ADSNP system using 93 neurons and astrocytes is given. The superiority of the ADSNP system is demonstrated by comparison with the six variants. Finally, an ADSNP system is constructed for credit card fraud detection, which verifies the feasibility of the ADSNP system for solving real-world problems. By considering the adaptive synaptic delay, ADSNP systems better restore the process of information transmission in biological neural networks, and enhance the adaptability of SNP systems, making the control of time more accurate.


Asunto(s)
Astrocitos , Modelos Neurológicos , Redes Neurales de la Computación , Neuronas , Sinapsis , Transmisión Sináptica , Sinapsis/fisiología , Astrocitos/fisiología , Neuronas/fisiología , Transmisión Sináptica/fisiología , Potenciales de Acción/fisiología , Adenosina Trifosfato/metabolismo , Factores de Tiempo , Humanos
5.
Commun Biol ; 7(1): 569, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750228

RESUMEN

Accumulation of amyloid-ß (Aß) and tau tangles are hallmarks of Alzheimer's disease. Aß is extracellular while tau tangles are typically intracellular, and it is unknown how these two proteinopathies are connected. Here, we use data of 1206 elders and test that RNA expression levels of GPER1, a transmembrane protein, modify the association of Aß with tau tangles. GPER1 RNA expression is related to more tau tangles (p = 0.001). Moreover, GPER1 expression modifies the association of immunohistochemistry-derived Aß load with tau tangles (p = 0.044). Similarly, GPER1 expression modifies the association between Aß proteoforms and tau tangles: total Aß protein (p = 0.030) and Aß38 peptide (p = 0.002). Using single nuclei RNA-seq indicates that GPER1 RNA expression in astrocytes modifies the relation of Aß load with tau tangles (p = 0.002), but not GPER1 in excitatory neurons or endothelial cells. We conclude that GPER1 may be a link between Aß and tau tangles driven mainly by astrocytic GPER1 expression.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Receptores de Estrógenos , Receptores Acoplados a Proteínas G , Proteínas tau , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Femenino , Masculino , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/genética , Anciano , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Anciano de 80 o más Años , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Astrocitos/metabolismo
6.
Neuropharmacology ; 253: 109982, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38701943

RESUMEN

Perioperative neurocognitive disorders (PND) are cognitive dysfunctions that usually occur in elderly patients after anesthesia and surgery. Microglial overactivation is a key underlying mechanism. Interleukin-33 (IL-33) is a member of the IL-1 family that orchestrates microglial function. In the present study, we explored how IL-33, which regulates microglia, contributes to cognitive improvement in a male mouse model of PND. An exploratory laparotomy was performed to establish a PND model. The expression levels of IL-33 and its receptor ST2 were evaluated using Western blot. IL-33/ST2 secretion, microglial density, morphology, phagocytosis of synapse, and proliferation, and dystrophic microglia were assessed using immunofluorescence. Synaptic plasticity was measured using Golgi staining and long-term potentiation. The Morris water maze and open field test were used to evaluate cognitive function and anxiety. Hippocampal expression of IL-33 and ST2 were elevated on postoperative day 3. We confirmed that IL-33 was secreted by astrocytes and neurons, whereas ST2 mainly colocalized with microglia. IL-33 treatment induced microgliosis after anesthesia and surgery. These microglia had larger soma sizes and shorter and fragmented branches. Compared to the Surgery group, IL-33 treatment reduced the synaptic phagocytosis of microglia and increased microglial proliferation and dystrophic microglia. IL-33 treatment also reversed the impaired synaptic plasticity and cognitive function caused by anesthesia and surgery. In conclusion, these results indicate that IL-33 plays a key role in regulating microglial state and synaptic phagocytosis in a PND mouse model. IL-33 treatment has a therapeutic potential for improving cognitive dysfunction in PND.


Asunto(s)
Interleucina-33 , Ratones Endogámicos C57BL , Microglía , Animales , Microglía/efectos de los fármacos , Microglía/metabolismo , Interleucina-33/metabolismo , Masculino , Ratones , Plasticidad Neuronal/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Complicaciones Cognitivas Postoperatorias/metabolismo , Fagocitosis/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Trastornos Neurocognitivos/metabolismo , Trastornos Neurocognitivos/tratamiento farmacológico , Modelos Animales de Enfermedad , Neuronas/efectos de los fármacos , Neuronas/metabolismo
7.
CNS Neurosci Ther ; 30(5): e14726, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38715251

RESUMEN

AIMS: The preoptic area (POA) of the hypothalamus, crucial in thermoregulation, has long been implicated in the pain process. However, whether nociceptive stimulation affects body temperature and its mechanism remains poorly studied. METHODS: We used capsaicin, formalin, and surgery to induce acute nociceptive stimulation and monitored rectal temperature. Optical fiber recording, chemical genetics, confocal imaging, and pharmacology assays were employed to confirm the role and interaction of POA astrocytes and extracellular adenosine. Immunofluorescence was utilized for further validation. RESULTS: Acute nociception could activate POA astrocytes and induce a decrease in body temperature. Manipulation of astrocytes allowed bidirectional control of body temperature. Furthermore, acute nociception and astrocyte activation led to increased extracellular adenosine concentration within the POA. Activation of adenosine A1 or A2A receptors contributed to decreased body temperature, while inhibition of these receptors mitigated the thermo-lowering effect of astrocytes. CONCLUSION: Our results elucidate the interplay between acute nociception and thermoregulation, specifically highlighting POA astrocyte activation. This enriches our understanding of physiological responses to painful stimuli and contributes to the analysis of the anatomical basis involved in the process.


Asunto(s)
Astrocitos , Hipotermia , Nocicepción , Área Preóptica , Animales , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Nocicepción/fisiología , Hipotermia/inducido químicamente , Masculino , Ratones , Receptores Purinérgicos P1/metabolismo , Ratones Endogámicos C57BL , Adenosina/metabolismo , Capsaicina/farmacología , Formaldehído/toxicidad , Formaldehído/farmacología
8.
Neurochem Int ; 177: 105765, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38750960

RESUMEN

BACKGROUND: Perioperative neurocognitive disorders (PND) are common complications after surgery in older patients. However, the specific mechanism of this condition remains unclear. Glial cell line-derived neurotrophic factor (GDNF) is an important neurotrophin that abundantly expressed throughout the brain. It can enhance synaptic plasticity and alleviate learning and memory impairments. Thus, the purpose of this study was to investigate the role of GDNF in PND and the mechanisms involved. METHODS: The PND animal model was established by performing left tibial fracture surgery on 18-month-old C57BL/6 mice under sevoflurane anesthesia. Recombinant adeno-associated virus (rAAV)-GDNF or empty vectors were injected bilaterally into the hippocampal CA1 region of aged mice 3 weeks before anesthesia/surgery. The open field and fear conditioning test were used to assess the behavior changes. Golgi staining and electrophysiology were utilized to evaluate the morphological and functional alterations of neuronal synaptic plasticity. Western blot analysis was carried out to measure the proteins expression levels and immunofluorescence staining was performed to probe the cellular localization of GDNF. RESULTS: Mice with surgery and anesthesia showed a significant decrease in hippocampus-dependent learning and memory, accompanied by a decline in hippocampal synaptic plasticity. Anesthesia/surgery induced a reduction of GDNF, which was colocalized with astrocytes. Overexpression of GDNF in astrocytes could ameliorate the decline in cognitive function by improving hippocampal synaptic plasticity, meanwhile astrocytic GDNF rescued the anesthesia/surgery-induced decrease in GFRα1 and NCAM. CONCLUSION: The study concludes that astrocytic GDNF may improve anesthesia/surgery-induced cognitive impairment by promoting hippocampal synaptic plasticity in aged mice via the GFRα1/NCAM pathway.


Asunto(s)
Astrocitos , Disfunción Cognitiva , Factor Neurotrófico Derivado de la Línea Celular Glial , Hipocampo , Ratones Endogámicos C57BL , Plasticidad Neuronal , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Plasticidad Neuronal/fisiología , Plasticidad Neuronal/efectos de los fármacos , Ratones , Astrocitos/metabolismo , Masculino , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Envejecimiento , Anestesia
9.
J Neurodev Disord ; 16(1): 27, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783199

RESUMEN

BACKGROUND: Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene and dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB). METHODS: We generated TSC disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies. RESULTS: Using microphysiological systems, we demonstrate that a BBB generated from TSC2 heterozygous mutant cells shows increased permeability. This can be rescued by wild type astrocytes or by treatment with rapamycin, an mTOR kinase inhibitor. CONCLUSION: Our results demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of cell lineages contributing to TSC pathogenesis and informs future therapeutics.


Asunto(s)
Barrera Hematoencefálica , Células Madre Pluripotentes Inducidas , Proteína 2 del Complejo de la Esclerosis Tuberosa , Esclerosis Tuberosa , Esclerosis Tuberosa/fisiopatología , Esclerosis Tuberosa/genética , Humanos , Barrera Hematoencefálica/fisiopatología , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Sirolimus/farmacología , Astrocitos/metabolismo
10.
Cell Death Dis ; 15(5): 361, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796462

RESUMEN

Disease models of neurodegeneration with brain iron accumulation (NBIA) offer the possibility to explore the relationship between iron dyshomeostasis and neurodegeneration. We analyzed hiPS-derived astrocytes from PANK2-associated neurodegeneration (PKAN), an NBIA disease characterized by progressive neurodegeneration and high iron accumulation in the globus pallidus. Previous data indicated that PKAN astrocytes exhibit alterations in iron metabolism, general impairment of constitutive endosomal trafficking, mitochondrial dysfunction and acquired neurotoxic features. Here, we performed a more in-depth analysis of the interactions between endocytic vesicles and mitochondria via superresolution microscopy experiments. A significantly lower number of transferrin-enriched vesicles were in contact with mitochondria in PKAN cells than in control cells, confirming the impaired intracellular fate of cargo endosomes. The investigation of cytosolic and mitochondrial iron parameters indicated that mitochondrial iron availability was substantially lower in PKAN cells compared to that in the controls. In addition, PKAN astrocytes exhibited defects in tubulin acetylation/phosphorylation, which might be responsible for unregulated vesicular dynamics and inappropriate iron delivery to mitochondria. Thus, the impairment of iron incorporation into these organelles seems to be the cause of cell iron delocalization, resulting in cytosolic iron overload and mitochondrial iron deficiency, triggering mitochondrial dysfunction. Overall, the data elucidate the mechanism of iron accumulation in CoA deficiency, highlighting the importance of mitochondrial iron deficiency in the pathogenesis of disease.


Asunto(s)
Astrocitos , Citosol , Sobrecarga de Hierro , Hierro , Mitocondrias , Astrocitos/metabolismo , Astrocitos/patología , Humanos , Mitocondrias/metabolismo , Citosol/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Tubulina (Proteína)/metabolismo , Fosforilación , Deficiencias de Hierro , Acetilación
11.
Biomed Pharmacother ; 175: 116739, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759288

RESUMEN

BACKGROUND: Ketamine, as a non-competitive antagonist of N-methyl-D-aspartate (NMDA) receptors, was originally used in general anesthesia. Epidemiological data show that ketamine has become one of the most commonly abused drugs in China. Ketamine administration might cause cognitive impairment; however, its molecular mechanism remains unclear. The glymphatic system is a lymphoid system that plays a key role in metabolic waste removal and cognitive regulation in the central nervous system. METHODS: Focusing on the glymphatic system, this study evaluated the behavioral performance and circulatory function of the glymphatic system by building a short-term ketamine administration model in mice, and detected the expression levels of the 5-HT2c receptor, ΔFosb, Pten, Akt, and Aqp4 in the hippocampus. Primary astrocytes were cultured to verify the regulatory relationships among related indexes using a 5-HT2c receptor antagonist, a 5-HT2c receptor short interfering RNA (siRNA), and a ΔFosb siRNA. RESULTS: Ketamine administration induced ΔFosb accumulation by increasing 5-HT2c receptor expression in mouse hippocampal astrocytes and primary astrocytes. ΔFosb acted as a transcription factor to recognize the AATGATTAAT bases in the 5' regulatory region of the Aqp4 gene (-1096 bp to -1087 bp), which inhibited Aqp4 expression, thus causing the circulatory dysfunction of the glymphatic system, leading to cognitive impairment. CONCLUSIONS: Although this regulatory mechanism does not involve the Pten/Akt pathway, this study revealed a new mechanism of ketamine-induced cognitive impairment in non-neuronal systems, and provided a theoretical basis for the safety of clinical treatment and the effectiveness of withdrawal.


Asunto(s)
Astrocitos , Disfunción Cognitiva , Sistema Glinfático , Hipocampo , Ketamina , Animales , Ketamina/farmacología , Ketamina/toxicidad , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Ratones , Masculino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Sistema Glinfático/efectos de los fármacos , Sistema Glinfático/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Acuaporina 4/metabolismo , Acuaporina 4/genética , Receptor de Serotonina 5-HT2C/metabolismo , Receptor de Serotonina 5-HT2C/genética , Ratones Endogámicos C57BL , Células Cultivadas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética
12.
Nat Commun ; 15(1): 4347, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773146

RESUMEN

Epigenetic mechanisms bridge genetic and environmental factors that contribute to the pathogenesis of major depression disorder (MDD). However, the cellular specificity and sensitivity of environmental stress on brain epitranscriptomics and its impact on depression remain unclear. Here, we found that ALKBH5, an RNA demethylase of N6-methyladenosine (m6A), was increased in MDD patients' blood and depression models. ALKBH5 in astrocytes was more sensitive to stress than that in neurons and endothelial cells. Selective deletion of ALKBH5 in astrocytes, but not in neurons and endothelial cells, produced antidepressant-like behaviors. Astrocytic ALKBH5 in the mPFC regulated depression-related behaviors bidirectionally. Meanwhile, ALKBH5 modulated glutamate transporter-1 (GLT-1) m6A modification and increased the expression of GLT-1 in astrocytes. ALKBH5 astrocyte-specific knockout preserved stress-induced disruption of glutamatergic synaptic transmission, neuronal atrophy and defective Ca2+ activity. Moreover, enhanced m6A modification with S-adenosylmethionine (SAMe) produced antidepressant-like effects. Our findings indicate that astrocytic epitranscriptomics contribute to depressive-like behaviors and that astrocytic ALKBH5 may be a therapeutic target for depression.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB , Astrocitos , Trastorno Depresivo Mayor , Ratones Noqueados , Animales , Astrocitos/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Ratones , Humanos , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/patología , Masculino , Femenino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Neuronas/metabolismo , Estrés Psicológico/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Conducta Animal , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Depresión/metabolismo , Depresión/genética , Adulto , Transmisión Sináptica , Persona de Mediana Edad
13.
Sci Rep ; 14(1): 11973, 2024 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796617

RESUMEN

The biology underlying proton minibeam radiation therapy (pMBRT) is not fully understood. Here we aim to elucidate the biological effects of pMBRT using Fourier Transform Infrared Microspectroscopy (FTIRM). In vitro (CTX-TNA2 astrocytes and F98 glioma rat cell lines) and in vivo (healthy and F98-bearing Fischer rats) irradiations were conducted, with conventional proton radiotherapy and pMBRT. FTIRM measurements were performed at ALBA Synchrotron, and multivariate data analysis methods were employed to assess spectral differences between irradiation configurations and doses. For astrocytes, the spectral regions related to proteins and nucleic acids were highly affected by conventional irradiations and the high-dose regions of pMBRT, suggesting important modifications on these biomolecules. For glioma, pMBRT had a great effect on the nucleic acids and carbohydrates. In animals, conventional radiotherapy had a remarkable impact on the proteins and nucleic acids of healthy rats; analysis of tumour regions in glioma-bearing rats suggested major nucleic acid modifications due to pMBRT.


Asunto(s)
Glioma , Terapia de Protones , Ratas Endogámicas F344 , Sincrotrones , Animales , Ratas , Glioma/radioterapia , Glioma/patología , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Línea Celular Tumoral , Astrocitos/efectos de la radiación , Astrocitos/metabolismo , Ácidos Nucleicos/efectos de la radiación , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo
14.
Viruses ; 16(5)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38793666

RESUMEN

SARS-CoV-2 primarily infects the lungs via the ACE2 receptor but also other organs including the kidneys, the gastrointestinal tract, the heart, and the skin. SARS-CoV-2 also infects the brain, but the hematogenous route of viral entry to the brain is still not fully characterized. Understanding how SARS-CoV-2 traverses the blood-brain barrier (BBB) as well as how it affects the molecular functions of the BBB are unclear. In this study, we investigated the roles of the receptors ACE2 and DPP4 in the SARS-CoV-2 infection of the discrete cellular components of a transwell BBB model comprising HUVECs, astrocytes, and pericytes. Our results demonstrate that direct infection on the BBB model does not modulate paracellular permeability. Also, our results show that SARS-CoV-2 utilizes clathrin and caveolin-mediated endocytosis to traverse the BBB, resulting in the direct infection of the brain side of the BBB model with a minimal endothelial infection. In conclusion, the BBB is susceptible to SARS-CoV-2 infection in multiple ways, including the direct infection of endothelium, astrocytes, and pericytes involving ACE2 and/or DPP4 and the blood-to-brain transcytosis, which is an event that does not require the presence of host receptors.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Astrocitos , Barrera Hematoencefálica , COVID-19 , Dipeptidil Peptidasa 4 , Pericitos , SARS-CoV-2 , Transcitosis , Internalización del Virus , Barrera Hematoencefálica/virología , Barrera Hematoencefálica/metabolismo , Humanos , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Pericitos/virología , Pericitos/metabolismo , COVID-19/virología , COVID-19/metabolismo , Astrocitos/virología , Astrocitos/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Encéfalo/virología , Encéfalo/metabolismo , Endocitosis , Células Endoteliales de la Vena Umbilical Humana/virología , Permeabilidad
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167211, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701957

RESUMEN

The interaction between glioma cells and astrocytes promotes the proliferation of gliomas. Micro-RNAs (miRNAs) carried by astrocyte exosomes (exos) may be involved in this process, but the mechanism remains unclear. The oligonucleotide AS1411, which consists of 26 bases and has a G-quadruplex structure, is an aptamer that targets nucleolin. In this study, we demonstrate exosome-miRNA-27a-mediated cross-activation between astrocytes and glioblastoma and show that AS1411 reduces astrocytes' pro-glioma activity. The enhanced affinity of AS1411 toward nucleolin is attributed to its G-quadruplex structure. After binding to nucleolin, AS1411 inhibits the entry of the NF-κB pathway transcription factor P65 into the nucleus, then downregulates the expression of miRNA-27a in astrocytes surrounding gliomas. Then, AS1411 downregulates astrocyte exosome-miRNA-27a and upregulates the expression of INPP4B, the target gene of miRNA-27a in gliomas, thereby inhibiting the PI3K/AKT pathway and inhibiting glioma proliferation. These results were verified in mouse orthotopic glioma xenografts and human glioma samples. In conclusion, the parallel structure of AS1411 allows it to bind to nucleolin and disrupt the exosome-miRNA-27a-mediated reciprocal activation loop between glioma cells and astrocytes. Our results may help in the development of a novel approach to therapeutic modulation of the glioma microenvironment.


Asunto(s)
Aptámeros de Nucleótidos , Astrocitos , Exosomas , Glioma , MicroARNs , Nucleolina , Oligodesoxirribonucleótidos , Fosfoproteínas , Proteínas de Unión al ARN , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Humanos , Astrocitos/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , MicroARNs/genética , MicroARNs/metabolismo , Animales , Glioma/metabolismo , Glioma/patología , Glioma/genética , Ratones , Oligodesoxirribonucleótidos/genética , Oligodesoxirribonucleótidos/metabolismo , Oligodesoxirribonucleótidos/farmacología , Aptámeros de Nucleótidos/metabolismo , Aptámeros de Nucleótidos/genética , Exosomas/metabolismo , Exosomas/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Transducción de Señal
16.
J Mater Chem B ; 12(21): 5085-5097, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38713059

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment associated with the accumulation of beta-amyloid protein (Aß). Aß activates glial cells in the brain, increasing the secretion of proinflammatory cytokines, which leads to neuroinflammation and neuronal death. Currently, there are no effective treatments that cure or stop its progression; therefore, AD is considered a global health priority. The main limitations are the low drug bioavailability and impermeability of the blood-brain barrier (BBB). Fortunately, nanomedicine has emerged as a promising field for the development of new nanosystems for the controlled and targeted delivery of drugs to the brain. Therefore, in this work, lipid-polymer hybrid nanoparticles (LPHNPs) conjugated with transferrin (Tf) to facilitate crossing the BBB and loaded with N-acetylcysteine (NAC) for its anti-inflammatory effect were synthesized, and their physicochemical characterization was carried out. Subsequently, an in vitro model involving human astrocytes derived from induced pluripotent stem cells (iPSC) from an AD-diagnosed patient was developed, which was brought to a reactive state by stimulation with lipopolysaccharides (LPSs). The cell culture was treated with either Tf-conjugated LPHNPs loaded with NAC (NAC-Tf-LPHNPs) at 0.25 mg mL-1, or free NAC at 5 mM. The results showed that NAC-Tf-LPHNPs favorably modulated the expression of proinflammatory genes such as interleukin-1ß (IL-1ß), amyloid precursor protein (APP) and glial fibrillary acidic protein (GFAP). In addition, they reduced the secretion of the proinflammatory cytokines interleukin 6 (IL-6), IL-1ß and interferon-gamma (INF-γ). Results for both cases were compared to the group of cells that did not receive any treatment. In contrast, free NAC only had this effect on the expression of IL-1ß and the secretion of the cytokines IL-6 and INF-γ. These results indicate the potential of NAC-Tf-LPHNPs for AD treatment.


Asunto(s)
Acetilcisteína , Enfermedad de Alzheimer , Astrocitos , Células Madre Pluripotentes Inducidas , Nanopartículas , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Nanopartículas/química , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Acetilcisteína/química , Acetilcisteína/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Polímeros/química , Polímeros/farmacología , Lípidos/química , Biomarcadores/metabolismo , Tamaño de la Partícula , Enfermedades Neuroinflamatorias/tratamiento farmacológico
17.
Ecotoxicol Environ Saf ; 279: 116480, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38772146

RESUMEN

Microcystins (MCs) are toxic to the central nervous system of mammals. However, the direct toxicity of MCs on mammalian brain cells and the involved molecular mechanisms are not fully elucidated. Here, we incubated primary astrocytes, the major glial cell-type in the brain, with 0-12.5 µM concentrations of MC-LR for 48 h, and the impairment was evaluated. We found that MC-LR caused significant increases in the cell viability at the range of 0.05-1 µM concentrations with the highest density at 0.1 µM concentration. Treatment with 0.1 µM MC-LR induced YAP nuclear translocation and decreased the ratio of p-YAP to YAP. It also decreased mRNA levels of the upstream regulator (AMOT), and enhanced expressions of YAP interacted genes (Egfr, Tead1, and Ctgf) in primary astrocytes. Overexpression of AMOT significantly attenuated the increase of MC-LR-induced astrocyte proliferation and the expression of YAP downstream genes. These results indicate that Hippo signaling contributed to MC-LR-caused astrocyte proliferation. Further, reactive astrogliosis was observed in the mice brain after MC-LR exposure to environmentally relevant concentrations (20 or 100 µg/L) through drinking water for 16 weeks. Pathological observations revealed that 100 µg/L MC-LR exposure caused neuronal damages with characteristics of shrunken or vacuolation in the region of the cerebral cortex, striatum and cerebellum. These results were accompanied with increased oxidative stress and inflammatory response. Our data reveal the potential astrocytic mechanisms in MC-induced neurotoxicity and raise an alarm for neurodegenerative disease risk following daily exposure to MC-LR.


Asunto(s)
Astrocitos , Proliferación Celular , Vía de Señalización Hippo , Toxinas Marinas , Microcistinas , Transducción de Señal , Microcistinas/toxicidad , Animales , Astrocitos/efectos de los fármacos , Vía de Señalización Hippo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Señalizadoras YAP , Supervivencia Celular/efectos de los fármacos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Receptores ErbB/metabolismo , Factores de Transcripción de Dominio TEA , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética
18.
PLoS Negl Trop Dis ; 18(5): e0012199, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38776344

RESUMEN

BACKGROUND: In Chagas disease (CD), a neglected tropical disease caused by the parasite Trypanosoma cruzi, the development of mental disorders such as anxiety, depression, and memory loss may be underpinned by social, psychological, and biological stressors. Here, we investigated biological factors underlying behavioral changes in a preclinical model of CD. METHODOLOGY/PRINCIPAL FINDINGS: In T. cruzi-infected C57BL/6 mice, a kinetic study (5 to 150 days postinfection, dpi) using standardized methods revealed a sequential onset of behavioral changes: reduced innate compulsive behavior, followed by anxiety and depressive-like behavior, ending with progressive memory impairments. Hence, T. cruzi-infected mice were treated (120 to 150 dpi) with 10 mg/Kg/day of the selective serotonin reuptake inhibitor fluoxetine (Fx), an antidepressant that favors neuroplasticity. Fx therapy reversed the innate compulsive behavior loss, anxiety, and depressive-like behavior while preventing or reversing memory deficits. Biochemical, histological, and parasitological analyses of the brain tissue showed increased levels of the neurotransmitters GABA/glutamate and lipid peroxidation products and decreased expression of brain-derived neurotrophic factor in the absence of neuroinflammation at 150 dpi. Fx therapy ameliorated the neurochemical changes and reduced parasite load in the brain tissue. Next, using the human U-87 MG astroglioma cell line, we found no direct effect of Fx on parasite load. Crucially, serotonin/5-HT (Ser/5-HT) promoted parasite uptake, an effect increased by prior stimulation with IFNγ and TNF but abrogated by Fx. Also, Fx blocked the cytokine-driven Ser/5-HT-promoted increase of nitric oxide and glutamate levels in infected cells. CONCLUSION/SIGNIFICANCE: We bring the first evidence of a sequential onset of behavioral changes in T. cruzi-infected mice. Fx therapy improves behavioral and biological changes and parasite control in the brain tissue. Moreover, in the central nervous system, cytokine-driven Ser/5-HT consumption may favor parasite persistence, disrupting neurotransmitter balance and promoting a neurotoxic environment likely contributing to behavioral and cognitive disorders.


Asunto(s)
Astrocitos , Enfermedad de Chagas , Fluoxetina , Ratones Endogámicos C57BL , Serotonina , Trypanosoma cruzi , Animales , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/psicología , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/fisiología , Serotonina/metabolismo , Ratones , Astrocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Encéfalo/efectos de los fármacos , Encéfalo/parasitología , Encéfalo/metabolismo , Conducta Animal/efectos de los fármacos , Masculino , Humanos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Cognición/efectos de los fármacos , Depresión/tratamiento farmacológico , Carga de Parásitos , Ansiedad/tratamiento farmacológico
19.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 204-208, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38814215

RESUMEN

Parkinson's disease (PD) remains the most common neurodegenerative disease worldwide, seriously affecting the normal life of patients. Currently, there is no effective clinical cure for PD. In this study, the research team explored the effect of ketamine (KET) on PD, which can lay a reliable foundation for future KET treatment of PD. First, the research team established a PD rat model with 6-hydroxydopamine (6-OHDA). The detection showed that the maximum angle of the inclined plate stay, the number of times of grid crossings and standing, and the ATPase activity in brain tissue were significantly lower in PD rats than in control rats, while the positive rate of α-synuclein in brain tissue was increased, showing typical pathological manifestations of PD. After using KET to intervene in PD rats, the behavioral and brain pathological changes were significantly alleviated, and the inflammation and oxidative stress damage of brain tissue were effectively reduced, suggesting the potential therapeutic effects of KET on PD. Furthermore, the use of KET inhibited the PI3K/AKT axis in the brain tissue of PD rats and promoted autophagy. Moreover, the significant suppression of the PI3K/AKT axis by KET was also demonstrated in the PD cell model established through lipopolysaccharide (LPS) inducement of astrocyte cell line HA1800. It is suggested that the mechanism of KET on PD is related to the inhibition of the PI3K/AKT axis.


Asunto(s)
Astrocitos , Ketamina , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Ratas Sprague-Dawley , Transducción de Señal , Animales , Ketamina/farmacología , Ketamina/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Masculino , Ratas , Autofagia/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Oxidopamina , Lipopolisacáridos , Humanos
20.
Neuropharmacology ; 255: 110001, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38750804

RESUMEN

Emerging evidence suggests an important role of astrocytes in mediating behavioral and molecular effects of commonly misused drugs. Passive exposure to nicotine alters molecular, morphological, and functional properties of astrocytes. However, a potential involvement of astrocytes in nicotine reinforcement remains largely unexplored. The overall hypothesis tested in the current study is that astrocytes play a critical role in nicotine reinforcement. Protein levels of the astrocyte marker glial fibrillary acidic protein (GFAP) were examined in key mesocorticolimbic regions following chronic nicotine intravenous self-administration. Fluorocitrate, a metabolic inhibitor of astrocytes, was tested for its effects on behaviors related to nicotine reinforcement and relapse. Effects of fluorocitrate on extracellular neurotransmitter levels, including glutamate, GABA, and dopamine, were determined with microdialysis. Chronic nicotine intravenous self-administration increased GFAP expression in the nucleus accumbens core (NACcr), but not other key mesocorticolimbic regions, compared to saline intravenous self-administration. Both intra-ventricular and intra-NACcr microinjection of fluorocitrate decreased nicotine self-administration. Intra-NACcr fluorocitrate microinjection also inhibited cue-induced reinstatement of nicotine seeking. Local perfusion of fluorocitrate decreased extracellular glutamate levels, elevated extracellular dopamine levels, but did not alter extracellular GABA levels in the NACcr. Fluorocitrate did not alter basal locomotor activity. These results indicate that nicotine reinforcement upregulates the astrocyte marker GFAP expression in the NACcr, metabolic inhibition of astrocytes attenuates nicotine reinforcement and relapse, and metabolic inhibition of astrocytes disrupts extracellular dopamine and glutamate transmission. Overall, these findings suggest that astrocytes play an important role in nicotine reinforcement and relapse, potentially through regulation of extracellular glutamate and dopamine neurotransmission.


Asunto(s)
Astrocitos , Citratos , Dopamina , Ácido Glutámico , Nicotina , Núcleo Accumbens , Ratas Wistar , Autoadministración , Animales , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Nicotina/farmacología , Nicotina/administración & dosificación , Masculino , Ácido Glutámico/metabolismo , Dopamina/metabolismo , Citratos/farmacología , Citratos/administración & dosificación , Ratas , Proteína Ácida Fibrilar de la Glía/metabolismo , Agonistas Nicotínicos/farmacología , Agonistas Nicotínicos/administración & dosificación , Microdiálisis , Refuerzo en Psicología , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA