Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Ther ; 31(5): 1313-1331, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-36739479

RESUMEN

Astrocyte-microglial interaction plays a crucial role in brain injury-associated neuroinflammation. Our previous data illustrated that astrocytes secrete microRNA, leading to anti-inflammatory effects on microglia. Long non-coding RNAs participate in neuroinflammation regulation after traumatic brain injury. However, the effect of astrocytes on microglial phenotype via long non-coding RNAs and the underlying molecular mechanisms remain elusive. We used long non-coding RNA sequencing on murine astrocytes and found that exosomal long non-coding RNA 4933431K23Rik attenuated traumatic brain injury-induced microglial activation in vitro and in vivo and ameliorated cognitive function deficiency. Furthermore, microRNA and messenger RNA sequencing together with binding prediction illustrated that exosomal long non-coding RNA 4933431K23Rik up-regulates E2F7 and TFAP2C expression by sponging miR-10a-5p. Additionally, E2F7 and TFAP2C, as transcription factors, regulated microglial Smad7 expression. Using Cx3cr1-Smad7 overexpression of adeno-associated virus, microglia specifically overexpressed Smad7 in the attenuation of neuroinflammation, resulting in less cognitive deficiency after traumatic brain injury. Mechanically, overexpressed Smad7 physically binds to IκBα and inhibits its ubiquitination, preventing NF-κB signaling activation. The Smad7 activator asiaticoside alleviates neuroinflammation and protects neuronal function in traumatic brain injury mice. This study revealed that an exosomal long non-coding RNA from astrocytes attenuates microglial activation after traumatic brain injury by up-regulating Smad7, providing a potential therapeutic target.


Asunto(s)
Lesiones Traumáticas del Encéfalo , MicroARNs , ARN Largo no Codificante , Ratones , Animales , Microglía/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Astrocitos/metabolismo , Enfermedades Neuroinflamatorias , MicroARNs/metabolismo , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/metabolismo , Fenotipo , Ratones Endogámicos C57BL
2.
Bull Environ Contam Toxicol ; 112(4): 51, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38556558

RESUMEN

Esketamine (ESK) is the S-enantiomer of ketamine racemate (a new psychoactive substance) that can result in illusions, and alter hearing, vision, and proprioception in human and mouse. Up to now, the neurotoxicity caused by ESK at environmental level in fish is still unclear. This work studied the effects of ESK on behaviors and transcriptions of genes in dopamine and GABA pathways in zebrafish larvae at ranging from 12.4 ng L- 1 to 11141.1 ng L- 1 for 7 days post fertilization (dpf). The results showed that ESK at 12.4 ng L- 1 significantly reduced the touch response of the larvae at 48 hpf. ESK at 12.4 ng L- 1 also reduced the time and distance of larvae swimming at the outer zone during light period, which implied that ESK might potentially decrease the anxiety level of larvae. In addition, ESK increased the transcription of th, ddc, drd1a, drd3 and drd4a in dopamine pathway. Similarly, ESK raised the transcription of slc6a1b, slc6a13 and slc12a2 in GABA pathway. This study suggested that ESK could affect the heart rate and behaviors accompanying with transcriptional alterations of genes in DA and GABA pathways at early-staged zebrafish, which resulted in neurotoxicity in zebrafish larvae.


Asunto(s)
Dopamina , Ketamina , Humanos , Animales , Ratones , Dopamina/metabolismo , Dopamina/farmacología , Pez Cebra/genética , Pez Cebra/metabolismo , Ketamina/metabolismo , Ketamina/farmacología , Larva , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
3.
Environ Sci Technol ; 57(36): 13384-13396, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37651267

RESUMEN

Imidacloprid (IMI) and thiamethoxam (THM) are ubiquitous in aquatic ecosystems. Their negative effects on parental fish are investigated while intergenerational effects at environmentally relevant concentrations remain unclear. In this study, F0 zebrafish exposed to IMI and THM (0, 50, and 500 ng L-1) for 144 days post-fertilization (dpf) was allowed to spawn with two modes (internal mating and cross-mating), resulting in four types of F1 generations to investigate the intergenerational effects. IMI and THM affected F0 zebrafish fecundity, gonadal development, sex hormone and VTG levels, with accumulations found in F0 muscles and ovaries. In F1 generation, paternal or maternal exposure to IMI and THM also influenced sex hormones levels and elevated the heart rate and spontaneous movement rate. LncRNA-mRNA network analysis revealed that cell cycle and oocyte meiosis-related pathways in IMI groups and steroid biosynthesis related pathways in THM groups were significantly enriched in F1 offspring. Similar transcriptional alterations of dmrt1, insl3, cdc20, ccnb1, dnd1, ddx4, cox4i1l, and cox5b2 were observed in gonads of F0 and F1 generations. The findings indicated that prolonged paternal or maternal exposure to IMI and THM could severely cause intergenerational toxicity, resulting in developmental toxicity and endocrine-disrupting effects in zebrafish offspring.


Asunto(s)
Exposición Materna , Pez Cebra , Animales , Femenino , Humanos , Tiametoxam , Ecosistema
4.
Bull Environ Contam Toxicol ; 110(1): 5, 2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36507940

RESUMEN

Agricultural use of neonicotinoid insecticides, neuroactive nitroguanidine compounds, has been detected everywhere in the global, posing significant hazard to nontarget organisms. This work studied the developmental neurotoxicity of zebrafish larvae exposed to imidacloprid (IMI) and thiamethoxam (THM), ranging from 0.05 µg L- 1 to 50 µg L- 1 for 35 days. Transcriptions of genes belonging to the behavior, neurodevelopment and cortisol synthesis in zebrafish larvae were monitored. The qPCR data demonstrated that with exposure time increased, the transcription of behavior related genes was down-regulated in both IMI and THM groups, such as macf1, cdh6 and syt10. Additionally, IMI and THM significantly up-regulated the transcriptions of actha, and down-regulated il1rapl1b and pi4k2a at 35 dpf. Importantly, IMI markedly enhanced the transcripiton of gfap, shha, nkx2.2a and nestin in a time dependent manner. This work provided the foundation for understanding zebrafish larvae's neurotoxicity induced by IMI and THM.


Asunto(s)
Insecticidas , Pez Cebra , Animales , Tiametoxam/toxicidad , Larva , Neonicotinoides/toxicidad , Nitrocompuestos/toxicidad , Insecticidas/toxicidad , Insecticidas/análisis
5.
Eur J Neurosci ; 53(4): 1350-1361, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33052619

RESUMEN

To explore the clinical characteristics and prognosis of COVID-19 patients with cerebral stroke. A total of 2,474 COVID-19 patients from February 10th to March 24th, 2020 were admitted and treated in two branches (Optic Valley and Sino-French New City branch) of the Tongji Hospital. Data on the clinical characteristics, laboratory parameters and prognosis of COVID-19 patients with or without cerebral stroke were collected and comparatively analysed. Of the 2,474 COVID-19 patients, 113 (4.7%) patients had cerebral stroke and 25 (1.0%) patients had new-onset stroke. Eighty-eight (77.9%) patients in the previous-stroke group had cerebral ischaemia, while 25 (22.1%) patients in the new-onset stroke group had cerebral ischaemia. Most COVID-19 patients with stroke were elderly with more comorbidities such as hypertension, diabetes and heart diseases than patients without stroke. Laboratory examinations showed hypercoagulation and elevated serum parameters such as IL-6, cTnI, NT pro-BNP and BUN. Consciousness disorders, a long disease course and poor prognosis were also more commonly observed in stroke patients. The mortality rate of stroke patients was almost double (12.4% vs. 6.9%) that of patients without stroke. In addition, age, male sex and hypertension were independent predictors for new cerebral stroke in COVID-19 patients. In conclusion, the high risk of new-onset stroke must be taken into consideration when treating COVID-19 patients with an elderly age combined with a history of hypertension. These patients are more vulnerable to multiorgan dysfunction and an overactivated inflammatory response, in turn leading to an unfavourable outcome and higher mortality rate.


Asunto(s)
COVID-19/complicaciones , Accidente Cerebrovascular/epidemiología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/mortalidad , Comorbilidad , Femenino , Humanos , Hipertensión/epidemiología , Incidencia , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , SARS-CoV-2 , Accidente Cerebrovascular/complicaciones
6.
J Neuroinflammation ; 17(1): 89, 2020 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-32192523

RESUMEN

BACKGROUND: The interaction between astrocytes and microglia plays a vital role in the damage and repair of brain lesions due to traumatic brain injury (TBI). Recent studies have shown that exosomes act as potent mediators involved in intercellular communication. METHODS: In the current study, the expression of inflammatory factors and miR-873a-5p in the lesion area and oedema area was evaluated in 15 patients with traumatic brain injury. Exosomes secreted by astrocytes were detected by immunofluorescence, Western blot and electron microscopy. A mouse model of TBI and an in vitro model of LPS-induced primary microglia were established to study the protective mechanism of exosomes from miR-873a-5p overexpressing in TBI-induced nerve injury. RESULTS: We discovered that exosomes derived from activated astrocytes promote microglial M2 phenotype transformation following TBI. More than 100 miRNAs were detected in these astrocyte-derived exosomes. miR-873a-5p is a major component that was highly expressed in human traumatic brain tissue. Moreover, miR-873a-5p significantly inhibited LPS-induced microglial M1 phenotype transformation and the subsequent inflammation through decreased phosphorylation of ERK and NF-κB p65. This effect also greatly improved the modified neurological severity score (mNSS) and attenuated brain injury in a strictly controlled cortical impact mouse model. CONCLUSIONS: Taken together, our research indicates that miRNAs in the exosomes derived from activated astrocytes play a key role in the astrocyte-microglia interaction. miR-873a-5p, as one of the main components of these astrocyte-derived exosomes, attenuated microglia-mediated neuroinflammation and improved neurological deficits following TBI by inhibiting the NF-κB signalling pathway. These findings suggest a potential role for miR-873a-5p in treating traumatic brain injury.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Exosomas/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Comunicación Celular/fisiología , Exosomas/química , Humanos , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo
7.
Tumour Biol ; 37(2): 2425-33, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26383521

RESUMEN

Cyclin-dependent kinase 4 (CDK4) is a member of cyclin-dependent kinase family which regulates G1 to S cell cycle transition. CDK4 activity is increased in many tumor types. Here, we report a negative automodulatory feedback loop between CDK4 and miR-16 that regulates cell cycle progression in nasopharyngeal carcinoma (NPC). By miRNA array and real-time PCR, we identified upregulation of tumor suppressor miR-16a, which inhibited cell cycle progression and sensitized NPC cells to chemotherapy. CDK4 knockdown reduced the expression of c-Myc, the latter of which directly suppresses the miR-16 expression by directly binding to the miR-16 promoter. Moreover, we found that miR-16 upregulation could reduce CDK4 expression by repressing CCND1 and thus forms a feedback loop via the CDK4/c-Myc/miR-16/CCND1 pathway. Finally, miR-16 was negatively correlated with CDK4 expression in NPC biopsies. In summary, our results define a double-negative feedback loop involving CDK4 and miR-16 mediated by c-Myc that modulates NPC cell growth and chemotherapy sensitivity.


Asunto(s)
Proliferación Celular/genética , Quinasa 4 Dependiente de la Ciclina/genética , MicroARNs/genética , Neoplasias Nasofaríngeas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Antineoplásicos/farmacología , Carcinoma , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen/métodos , Genes Supresores de Tumor/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
8.
BMC Cancer ; 16: 238, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26993269

RESUMEN

BACKGROUND: In previous investigation, we reported that stably knocking down cyclin-dependent kinase 4(CDK4) induced expression of let-7c, which further suppressed cell cycle transition and cell growth by modulating cell cycle signaling in nasopharyngeal carcinoma (NPC). In this study, we further explored the molecular function and mechanism of CDK4 modulating miRNAs to stimulate cell cycle transition, cell growth, and Cisplatin (DDP) -resistance on in NPC. METHODS: We identified changes in miRNAs by miRNA array and real-time PCR and the effect on DDP after knocking down CDK4 in NPC cells. Further, we investigated the molecular mechanisms by which CDK4 modulated miR-15a in NPC. Moreover, we also explored the role of miR-15a and the effect on DDP in NPC. Finally, we analyzed the correlation of miR-15a and CDK4 expression in NPC tissues. RESULTS: In addition to let-7 family members, we observed that upregulated expression of miR-15a was significantly induced in CDK4-suppressed NPC cells. Further, we found that knocking down CDK4 suppressed c-Myc expression, and the latter directly suppressed the expression of miR-15a in NPC. Furthermore, miR-15a as a tumor suppressor antagonized CDK4 repressing cell cycle progression and cell growth in vitro and in vivo and induced the sensitivity of cells to DDP by regulating the c-Myc/CCND1/CDK4/E2F1 pathway in NPC. Finally, miR-15a was negatively weak correlated with the expression of CDK4 in NPC. CONCLUSIONS: Our studies demonstrate that CDK4 and miR-15a comprise an abnormal automodulatory feedback loop stimulating the pathogenesis and inducing chemotherapy resistance in NPC.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , MicroARNs/metabolismo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/genética , Carcinoma , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Quinasa 4 Dependiente de la Ciclina/metabolismo , Resistencia a Antineoplásicos/genética , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Transducción de Señal/efectos de los fármacos
10.
Sci Total Environ ; 932: 172872, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38692322

RESUMEN

The misuse of antitussives preparations is a continuing problem in the world, and imply that they might have potential new psychoactive substances (NPS) activity. However, few study focus on their ecological toxicity towards fish. In the present study, the machine learning (ML) methods gcForest and random forest (RF) were employed to predict NPS activity in 30 antitussives. The potential toxic target, mode of action (MOA), acute toxicity and chronic toxicity to fish were further investigated. The results showed that both gcForest and RF achieved optimal performance when utilizing combined features of molecular fingerprint (MF) and molecular descriptor (MD), with area under the curve (AUC) = 0.99, accuracy >0.94 and f1 score > 0.94, and were applied to screen the NPS activity in antitussives. A total of 15 antitussives exhibited potential NPS activity, including frequently-used substances like codeine and dextromethorphan. The binding affinity of these antitussives with zebrafish dopamine transporter (zDAT) was high, and even surpassing that of some traditional narcotics and NPS. Some antitussives formed hydrogen bonds or salt bridges with aspartate (Asp) 95, tyrosine (Tyr) 171 of zDAT. For the ecotoxicity, the MOA of these 15 antitussives in fish was predicted as narcosis. The prenoxdiazin, pholcodine, codeine, dextromethorphan and dextrorphan exhibited very toxic/toxic to fish. It was necessary to pay close attention to the ecotoxicity of these antitussives. In this study, the integration of ML, molecular docking and ECOSAR approaches are powerful tools for understanding the toxicity profiles and ecological hazards posed by new pollutants.


Asunto(s)
Psicotrópicos , Contaminantes Químicos del Agua , Animales , Contaminantes Químicos del Agua/toxicidad , Psicotrópicos/toxicidad , Pez Cebra , Peces , Aprendizaje Automático
11.
Sci Total Environ ; 923: 171475, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38453063

RESUMEN

Climbazole is an azole biocide that has been widely used in formulations of personal care products. Climbazole can cause developmental toxicity and endocrine disruption as well as gut disturbance in aquatic organisms. However, the mechanisms behind gut toxicity induced by climbazole still remain largely unclear in fish. Here, we evaluate the gut effects by exposing grass carp (Ctenopharyngodon idella) to climbazole at levels ranging from 0.2 to 20 µg/L for 42 days by evaluating gene transcription and expression, biochemical analyses, correlation network analysis, and molecular docking. Results showed that climbazole exposure increased cyp1a mRNA expression and ROS level in the three treatment groups. Climbazole also inhibited Nrf2 and Keap1 transcripts as well as proteins, and suppressed the transcript levels of their subordinate antioxidant molecules (cat, sod, and ho-1), increasing oxidative stress. Additionally, climbazole enhanced NF-κB and iκBα transcripts and proteins, and the transcripts of NF-κB downstream pro-inflammatory factors (tnfα, and il-1ß/6/8), leading to inflammation. Climbazole increased pro-apoptosis-related genes (fadd, bad1, and caspase3), and decreased anti-apoptosis-associated genes (bcl2, and bcl-xl), suggesting a direct reaction to apoptosis. The molecular docking data showed that climbazole could form stable hydrogen bonds with CYP1A. Mechanistically, our findings suggested that climbazole can induce inflammation and oxidative stress through CYP450s/ROS/Nrf2/NF-κB pathways, resulting in cell apoptosis in the gut of grass carp.


Asunto(s)
Carpas , Suplementos Dietéticos , Imidazoles , Animales , Suplementos Dietéticos/análisis , Dieta , FN-kappa B , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Inmunidad Innata , Azoles/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Simulación del Acoplamiento Molecular , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Inflamación/inducido químicamente , Inflamación/veterinaria , Estrés Oxidativo , Apoptosis , Carpas/metabolismo
12.
J Hazard Mater ; 468: 133844, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38394900

RESUMEN

Androgen receptor (AR) agonists have strong endocrine disrupting effects in fish. Most studies mainly investigate AR binding capacity using human AR in vitro. However, there is still few methods to rapidly predict AR agonists in aquatic organisms. This study aimed to screen AR agonists of fish species using machine learning and molecular models in water-relevant list from NORMAN, a network of reference laboratories for monitoring contaminants of emerging concern in the environment. In this study, machine learning approaches (e.g., Deep Forest (DF)), Random Forests and artificial neural networks) were applied to predict AR agonists. Zebrafish, fathead minnow, mosquitofish, medaka fish and grass carp are all important aquatic model organisms widely used to evaluate the toxicity of new pollutants, and the molecular models of ARs from these five fish species were constructed to further screen AR agonists using AlphaFold2. The DF method showed the best performances with 0.99 accuracy, 0.97 sensitivity and 1 precision. The Asn705, Gln711, Arg752, and Thr877 residues in human AR and the corresponding sites in ARs from the five fish species were responsible for agonist binding. Overall, 245 substances were predicted as suspect AR agonists in the five fish species, including, certain glucocorticoids, cholesterol metabolites, and cardiovascular drugs in the NORMAN list. Using machine learning and molecular modeling hybrid methods rapidly and accurately screened AR agonists in fish species, and helping evaluate their ecological risk in fish populations.


Asunto(s)
Andrógenos , Disruptores Endocrinos , Peces , Receptores Androgénicos , Animales , Humanos , Andrógenos/química , Andrógenos/toxicidad , Cyprinidae , Aprendizaje Automático , Modelos Moleculares , Pez Cebra , Disruptores Endocrinos/química , Disruptores Endocrinos/toxicidad
13.
J Hazard Mater ; 465: 133463, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38219582

RESUMEN

Azole antifungal climbazole has frequently been detected in aquatic environments and shows various effects in fish. However, the underlying mechanism of toxicity through the gut-brain axis of climbazole is unclear. Here, we investigated the effects of climbazole at environmental concentrations on the microbiota-intestine-brain axis in grass carp via histopathological observation, gene expression and biochemical analyses, and high-throughput sequencing of the 16 S rRNA. Results showed that exposure to 0.2 to 20 µg/L climbazole for 42 days significantly disrupted gut microbiota and caused brain neurotoxicity in grass carp. In this study, there was an alteration in the phylum and genus compositions in the gut microbiota following climbazole treatment, including reducing Fusobacteria (e.g., Cetobacterium) and increasing Actinobacteria (e.g., Nocardia). Climbazole disrupted intestinal microbial abundance, leading to increased levels of lipopolysaccharide and tumor necrosis factor-alpha in the gut, serum, and brain. They passed through the impaired intestinal barrier into the circulation and caused the destruction of the blood-brain barrier through the gut-brain axis, allowing them into the brain. In the brain, climbazole activated the nuclear factor kappaB pathway to increase inflammation, and suppressed the E2-related factor 2 pathway to produce oxidative damage, resulting in apoptosis, which promoted neuroinflammation and neuronal death. Besides, our results suggested that this neurotoxicity was caused by the breakdown of the microbiota-gut-brain axis, mediated by reduced concentrations of dopamine, short chain fatty acids, and intestinal microbial activity induced by climbazole.


Asunto(s)
Carpas , Fungicidas Industriales , Imidazoles , Animales , Eje Cerebro-Intestino , Azoles
14.
Aquat Toxicol ; 261: 106604, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37311377

RESUMEN

Progestins are widely used and detected in surface waters, and can affect gonad development and sexual differentiation in fish. However, the toxicological mechanisms of sexual differentiation induced by progestins are not well understood. Here, we investigated the effects of norethindrone (NET) and androgen receptor (AR) antagonist flutamide (FLU) on gonadal differentiation in zebrafish from 21 dpf (days post-fertilization) to 49 dpf. The results showed that NET caused male bias, while FLU resulted in female bias at 49 dpf. The NET and FLU mixtures significantly decreased the percentage of males compared to the NET single exposure. Molecular docking analysis showed that FLU and NET had similar docking pocket and docking posture with AR resulting in competitively forming the hydrogen bond with Thr334 of AR. These results suggested that binding to AR was the molecular initiating event of sex differentiation induced by NET. Moreover, NET strongly decreased transcription of biomarker genes (dnd1, ddx4, dazl, piwil1 and nanos1) involved in germ cell development, while FLU significantly increased transcription of these target genes. There was an increase in the number of juvenile oocytes, which was consistent with the female bias in the combined groups. The bliss independence model analysis further showed that NET and FLU had antagonistic effect on transcription and histology during gonadal differentiation. Thus, NET suppressed the germ cell development via AR, resulting in male bias. Understanding the molecular initiation of sex differentiation in progestins is essential to provide a comprehensive biological basis for ecological risk assessment.


Asunto(s)
Noretindrona , Contaminantes Químicos del Agua , Animales , Masculino , Femenino , Noretindrona/farmacología , Progestinas/farmacología , Receptores Androgénicos , Pez Cebra/genética , Simulación del Acoplamiento Molecular , Contaminantes Químicos del Agua/toxicidad , Flutamida/toxicidad , Diferenciación Sexual , Células Germinativas , Diferenciación Celular
15.
Aquat Toxicol ; 265: 106765, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37979497

RESUMEN

Ephedrine (EPH) and cocaine (COC) are illegal stimulant drugs, and have been frequently detected in aquatic environments. EPH and COC have negative effects on the nervous system and cause abnormal behaviors in mammals and fish at high concentrations, but their mechanisms of neurotoxicity remain unclear in larvae fish at low concentrations. To address this issue, zebrafish embryos were exposed to EPH and COC for 14 days post-fertilization (dpf) at 10, 100, and 1000 ng L-1. The bioaccumulation, development, behavior, cell neurotransmitter levels and apoptosis were detected to investigate the developmental neurotoxicity (DNT) of EPH and COC. The results showed that EPH decreased heart rate, while COC increased heart rate. EPH caused cell apoptosis in the brain by AO staining. In addition, behavior analysis indicated that EPH and COC affected spontaneous movement, touch-response, swimming activity and anxiety-like behaviors. EPH and COC altered the levels of the neurotransmitters dopamine (DA) and γ-aminobutyric acid (GABA) with changes of the transcription of genes related to the DA and GABA pathways. These findings indicated that EPH and COC had noticeable DNT in the early stage of zebrafish at environmentally relevant concentrations.


Asunto(s)
Cocaína , Contaminantes Químicos del Agua , Animales , Pez Cebra/metabolismo , Efedrina/toxicidad , Efedrina/metabolismo , Contaminantes Químicos del Agua/toxicidad , Cocaína/toxicidad , Cocaína/metabolismo , Neurotransmisores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Larva , Mamíferos/metabolismo
16.
Aquat Toxicol ; 263: 106698, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37722153

RESUMEN

Climbazole, an azole, is widely used in personal care products, pharmaceuticals, and pesticides and is frequently detected in surface water. Climbazole has showed endocrine-disrupting effects. However, the effects of climbazole in fish are still largely unclear. In this study, grass carp (Ctenopharyngodon idella) and liver cell lines (L8824 cells) were treated with climbazole at concentrations ranging from 0.2 to 20 µg/L for 42 days in vivo and 24 h in vitro to evaluate the effects on the liver, respectively. Pathological, biochemical, and gene transcription and expression analyses were conducted to examine the hepatotoxicity. Our results showed that climbazole significantly decreased the hepatosomatic index, caused cell apoptosis in vivo and in vitro, and finally accumulated lipids in the liver. Beside, climbazole increased ROS levels, reduced Nrf2 and Keap1 mRNA and protein levels, and further decreased transcription of Nrf2-dependent downstream antioxidant enzyme genes, causing oxidative stress. Moreover, climbazole increased transcription and protein levels of apoptosis-related genes. Finally, climbazole damaged mitochondrial function and structure, disrupted liver lipid metabolism. Overall, climbazole caused hepatotoxicity, leading to a high ecological risk for aquatic organisms.

17.
Bioengineered ; 13(3): 4646-4657, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35148670

RESUMEN

To explore the effects of antiepileptic drug sodium valproate on magnetic resonance imaging (MRI) images, neurological cognition, and JAK1/STAT3 pathway in hippocampus of rats with depression, 30 Sprague Dawley (SD) rats were included. The depression model (DM) was prepared through the chronic stress restraint test. Some model rats were injected with 10 mg/kg sodium valproate into abdominal cavity before modeling (RT group)), and healthy rats were selected as controls (healthy control (HC) group). Depth of split brain was greatly increased in DM group, and nitrogen-acetyl aspartic acid (NAA)/creatine (Cr), glutamic acid (Glu)/Cr, and choline (Cho)/Cr ratios were greatly reduced (P < 0.05). Behavioral test results showed that sugar water preference rate, escape latency, and divergence index in DM group were greatly reduced (P < 0.05), and cumulative immobility time, target quadrant stay time, and number of crossings in forced swimming and tail suspension were prolonged dramatically (P < 0.05), with no difference between the two groups (P > 0.05). Expression levels of interleukin 1ß (IL-1ß) and interleukin 6 (IL-6) in hippocampus of DM group were obviously increased (P < 0.05), and expression levels of JAK1 and STAT3 were decreased visibly (P < 0.05), with no difference between the two (P > 0.05). In summary, anti-epileptic drug sodium valproate effectively improves hippocampal volume characteristics and memory and neurocognitive dysfunction of depression models.


Asunto(s)
Anticonvulsivantes , Depresión , Animales , Anticonvulsivantes/metabolismo , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Ácido Aspártico/metabolismo , Creatina/metabolismo , Depresión/tratamiento farmacológico , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Imagen por Resonancia Magnética , Ratas , Ratas Sprague-Dawley , Ácido Valproico/metabolismo
18.
Aquat Toxicol ; 248: 106177, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35512552

RESUMEN

Synthetic progestins levonorgestrel (LNG) and dydrogesterone (DDG) are frequency detected in surface water. Combined effects of LNG and DDG on gonad differentiation are similar to LNG single exposure in juvenile zebrafish. However, LNG and DDG mixtures have stronger effects on spermatogenesis in testes of adult zebrafish, which show variable at different life stage. Effects of LNG and DDG mixtures on eyes and brain remain unknown. Here we investigated effects of LNG, DDG and their mixtures on eyes and brain. Zebrafish were exposed to LNG, DDG and their mixtures from 2 hpf to 144 dpf. Rhythm and vision related biological processes were enriched in eyes and brain in LNG and DDG treatments, which indicated rhythmic oscillation in eyes and brain. The qPCR data revealed that both LNG and DDG decreased transcription of arntl2 and clocka, while increased transcription of per1a, per1b, rpe65a and tefa in eyes and brain. However, DDG and LNG mixtures had slight effect on transcription of genes related to rhythm and vision. In addition, LNG and DDG reduced the thickness of inner nuclear layer in the eyes. Bliss independent model revealed that LNG and DDG had antagonist effects on transcription and histology in eyes and brain. Moreover, LNG and DDG formed the same hydrogen bonds with green-sensitive opsin-4 and rhodopsin kinase GRK7a. Taken together, LNG and DDG competed with each other for the same binding residues resulting in antagonist effect in their mixtures treatments, and have significant ecological implications to assess combined effects of progestins mixtures on fish in different organs.


Asunto(s)
Didrogesterona , Contaminantes Químicos del Agua , Animales , Encéfalo/metabolismo , Levonorgestrel/toxicidad , Masculino , Proteínas Circadianas Period/metabolismo , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
19.
Int J Mol Med ; 49(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34751408

RESUMEN

Neuroinflammatory processes mediated by microglial activation and subsequent neuronal damage are the hallmarks of traumatic brain injury (TBI). As an inhibitor of the macrophage­inducible C­type lectin (Mincle)/spleen tyrosine kinase (Syk) signaling pathway, BAY61­3606 (BAY) has previously demonstrated anti­inflammatory effects on some pathological processes, such as acute kidney injury, by suppressing the inflammatory macrophage response. In the present study, the potential effects of BAY on microglial phenotype and neuroinflammation after TBI were investigated. BAY (3 mg/kg) was first administered into mice by intraperitoneal injection after TBI induction in vivo and microglia were also treated with BAY (2 µM) in vitro. The levels of inflammatory factors in microglia were assessed using reverse transcription­quantitative PCR and ELISA. Cortical neuron, myelin sheath, astrocyte and cerebrovascular endothelial cell markers were detected using immunofluorescence. The levels of components of the Mincle/Syk/NF­κB signaling pathway [Mincle, phosphorylated (p)­Syk and NF­κB], in addition to proteins associated with inflammation (ASC, caspase­1, TNF­α, IL­1ß and IL­6), apoptosis (Bax and Bim) and tight junctions (Claudin­5), were measured via western blotting and ELISA. Migration and chemotaxis of microglial cells were evaluated using Transwell and agarose spot assays. Neurological functions of the mice were determined in vivo using the modified neurological severity scoring system and a Morris water maze. The results of the present study revealed that the expression levels of proteins in the Mincle/Syk/NF­κB signaling pathway (including Mincle, p­Syk and p­NF­κB), inflammatory cytokines (TNF­α, IL­1ß and IL­6), proteins involved in inflammation (ASC and caspase­1), apoptotic markers (Bax and Bim) and the tight junction protein Claudin­5 were significantly altered post­TBI. BAY treatment reversed these effects in both the cerebral cortex extract­induced cell model and the controlled cortical impact mouse model. BAY was also revealed to suppress activation of the microglial proinflammatory phenotype and microglial migration. In addition, BAY effectively attenuated TBI­induced neurovascular unit damage and neurological function deficits. Taken together, these findings provided evidence that BAY may inhibit the Mincle/Syk/NF­κB signaling pathway in microglia; this in turn could attenuate microglia­mediated neuroinflammation and improve neurological deficits following TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lectinas Tipo C/metabolismo , Microglía/efectos de los fármacos , Niacinamida/análogos & derivados , Pirimidinas/farmacología , Receptores Inmunológicos/metabolismo , Quinasa Syk/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Estudios de Casos y Controles , Niño , Humanos , Masculino , Ratones Endogámicos C57BL , Microglía/patología , Persona de Mediana Edad , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/fisiopatología , Fármacos Neuroprotectores/farmacología , Niacinamida/farmacología , Células PC12 , Ratas , Adulto Joven
20.
Front Pharmacol ; 12: 719823, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34744713

RESUMEN

Background: Phillyrin (Phi) is the main polyphenolic compound found in Forsythia suspensa. Recent studies have revealed that Phi has potent antioxidative and anti-inflammatory effects. However, whether Phi could relieve blood-brain barrier (BBB) damage following traumatic brain injury (TBI) remains unknown. Materials and Methods: Lipopolysaccharide (LPS) was used to activate primary microglia, which were then treated with different doses of Phi or the peroxisome proliferator-activated receptor-gamma (PPARγ) antagonist (GW9662). CCK-8 assay was used for evaluating cell viability, and the cytokines (including IL-1ß, IL-6, TNFα, IL-4, IL-10, and TGFß), microglial phenotypic markers (iNOS, COX2, and CD86 for "M1" polarization; Arg1, Ym1, and CD206 for "M2" polarization), PPARγ, and NF-κB were determined by RT-PCR, Western blot, or cellular immunofluorescence. Primary cultured mouse brain microvascular endothelial cells (BMECs) were stimulated by the condition medium (CM) from microglia. The cell viability, angiogenesis, and tight junction of BMECs were determined via CCK-8 assay, tube formation assay, and Western blot (for detecting MMP3, MMP9, ZO1, claudin-5, and occludin). Furthermore, the mouse TBI model was constructed and treated with Phi and/or GW9662. The BBB integrity was evaluated by H&E staining, Evans blue staining, and tissue immunofluorescence. Results: Phi markedly restrained the pro-inflammatory ("M1" state) cytokines and promoted anti-inflammatory ("M2" polarization) cytokines in LPS-mediated microglia. Phi mitigated "M1" polarization and promoted "M2" polarization of microglia via enhancing PPARγ and inhibiting the NF-κB pathway. The PPARγ antagonist GW9662 significantly repressed Phi-mediated anti-inflammatory effects. Meanwhile, Phi enhanced the viability, tube formation ability, and cell junction of BMECs. In the TBI mouse model, Phi promoted "M2" polarization, whereas it repressed the "M1" polarization of microglia. In addition, Phi reduced TBI-mediated BBB damage. However, the protective effects of Phi were reversed mainly by GW9662 treatment. Conclusion: Phi prevents BBB damage via inhibiting the neuroinflammation of microglia through the PPARγ/NF-κB pathway, which provides a potential therapeutic drug against TBI.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA