Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 30, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38331979

RESUMO

Worldwide, the incidence of major depressive disorder (MDD) is increasing annually, resulting in greater economic and social burdens. Moreover, the pathological mechanisms of MDD and the mechanisms underlying the effects of pharmacological treatments for MDD are complex and unclear, and additional diagnostic and therapeutic strategies for MDD still are needed. The currently widely accepted theories of MDD pathogenesis include the neurotransmitter and receptor hypothesis, hypothalamic-pituitary-adrenal (HPA) axis hypothesis, cytokine hypothesis, neuroplasticity hypothesis and systemic influence hypothesis, but these hypothesis cannot completely explain the pathological mechanism of MDD. Even it is still hard to adopt only one hypothesis to completely reveal the pathogenesis of MDD, thus in recent years, great progress has been made in elucidating the roles of multiple organ interactions in the pathogenesis MDD and identifying novel therapeutic approaches and multitarget modulatory strategies, further revealing the disease features of MDD. Furthermore, some newly discovered potential pharmacological targets and newly studied antidepressants have attracted widespread attention, some reagents have even been approved for clinical treatment and some novel therapeutic methods such as phototherapy and acupuncture have been discovered to have effective improvement for the depressive symptoms. In this work, we comprehensively summarize the latest research on the pathogenesis and diagnosis of MDD, preventive approaches and therapeutic medicines, as well as the related clinical trials.


Assuntos
Transtorno Depressivo Maior , Humanos , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/genética , Transtorno Depressivo Maior/prevenção & controle , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal
2.
Neural Regen Res ; 19(5): 1126-1133, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-37862218

RESUMO

Traumatic spinal cord injury is potentially catastrophic and can lead to permanent disability or even death. China has the largest population of patients with traumatic spinal cord injury. Previous studies of traumatic spinal cord injury in China have mostly been regional in scope; national-level studies have been rare. To the best of our knowledge, no national-level study of treatment status and economic burden has been performed. This retrospective study aimed to examine the epidemiological and clinical features, treatment status, and economic burden of traumatic spinal cord injury in China at the national level. We included 13,465 traumatic spinal cord injury patients who were injured between January 2013 and December 2018 and treated in 30 hospitals in 11 provinces/municipalities representing all geographical divisions of China. Patient epidemiological and clinical features, treatment status, and total and daily costs were recorded. Trends in the percentage of traumatic spinal cord injuries among all hospitalized patients and among patients hospitalized in the orthopedic department and cost of care were assessed by annual percentage change using the Joinpoint Regression Program. The percentage of traumatic spinal cord injuries among all hospitalized patients and among patients hospitalized in the orthopedic department did not significantly change overall (annual percentage change, -0.5% and 2.1%, respectively). A total of 10,053 (74.7%) patients underwent surgery. Only 2.8% of patients who underwent surgery did so within 24 hours of injury. A total of 2005 (14.9%) patients were treated with high-dose (≥ 500 mg) methylprednisolone sodium succinate/methylprednisolone (MPSS/MP); 615 (4.6%) received it within 8 hours. The total cost for acute traumatic spinal cord injury decreased over the study period (-4.7%), while daily cost did not significantly change (1.0% increase). Our findings indicate that public health initiatives should aim at improving hospitals' ability to complete early surgery within 24 hours, which is associated with improved sensorimotor recovery, increasing the awareness rate of clinical guidelines related to high-dose MPSS/MP to reduce the use of the treatment with insufficient evidence.

3.
J Psychiatr Res ; 161: 188-198, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36933445

RESUMO

Posttraumatic stress disorder (PTSD) is very common after exposure to trauma, mental stress or violence. Because objective biological markers for PTSD are lacking, exactly diagnosing PTSD is a challenge for clinical psychologists. In-depth research on the pathogenesis of PTSD is a key for solving this problem. In this work, we used male Thy1-YFP transgenic mice, in which neurons are fluorescently labeled, to research the effects of PTSD on neurons in vivo. We initially discovered that pathological stress associated with PTSD increased the activation of glycogen synthesis kinase-beta (GSK-3ß) in neurons and induced the translocation of the transcription factor forkhead box-class O3a (FoxO3a) from the cytoplasm to the nucleus, which decreased the expression of uncoupling protein 2 (UCP2) and increased mitochondrial production of reactive oxygen species (ROS) to trigger neuronal apoptosis in the prefrontal cortex (PFC). Furthermore, the PTSD model mice showed increased freezing and anxiety-like behaviors and more severe decrease of memory and exploratory behavior. Additionally, leptin attenuated neuronal apoptosis by increasing the phosphorylation of signal transducer and activator of transcription 3 (STAT3), which further elevated the expression of UCP2 and inhibited the mitochondrial production of ROS induced by PTSD, thus reducing neuronal apoptosis and ameliorating PTSD-related behaviors. Our study is expected to promote the exploration of PTSD-related pathogenesis in neural cells and the clinical effectiveness of leptin for PTSD.


Assuntos
Transtornos de Estresse Pós-Traumáticos , Camundongos , Masculino , Animais , Transtornos de Estresse Pós-Traumáticos/metabolismo , Leptina , Camundongos Transgênicos , Espécies Reativas de Oxigênio , Glicogênio Sintase Quinase 3 beta
4.
Mol Psychiatry ; 28(7): 3044-3054, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36991130

RESUMO

Neuropathological mechanisms of manic syndrome or manic episodes in bipolar disorder remain poorly characterised, as the research progress is severely limited by the paucity of appropriate animal models. Here we developed a novel mania mice model by combining a series of chronic unpredictable rhythm disturbances (CURD), which include disruption of circadian rhythm, sleep deprivation, exposure to cone light, with subsequent interference of followed spotlight, stroboscopic illumination, high-temperature stress, noise disturbance and foot shock. Multiple behavioural and cell biology tests comparing the CURD-model with healthy controls and depressed mice were deployed to validate the model. The manic mice were also tested for the pharmacological effects of various medicinal agents used for treating mania. Finally, we compared plasma indicators of the CURD-model mice and the patients with the manic syndrome. The CURD protocol produced a phenotype replicating manic syndrome. Mice exposed to CURD presented manic behaviours similar to that observed in the amphetamine manic model. These behaviours were distinct from depressive-like behaviours recorded in mice treated with a depression-inducing protocol of chronic unpredictable mild restraint (CUMR). Functional and molecular indicators in the CURD mania model showed multiple similarities with patients with manic syndrome. Treatment with LiCl and valproic acid resulted in behavioural improvements and recovery of molecular indicators. A novel manic mice model induced by environmental stressors and free from genetic or pharmacological interventions is a valuable tool for research into pathological mechanisms of mania.


Assuntos
Transtorno Bipolar , Mania , Humanos , Animais , Camundongos , Modelos Animais de Doenças , Ácido Valproico , Privação do Sono
5.
Cell Death Dis ; 13(4): 406, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35468877

RESUMO

Alzheimer's disease (AD) is the prevalent cause of dementia in the ageing world population. Apolipoprotein E4 (ApoE4) allele is the key genetic risk factor for AD, although the mechanisms linking ApoE4 with neurocognitive impairments and aberrant metabolism remains to be fully characterised. We discovered a significant increase in the ApoE4 content of serum exosomes in old healthy subjects and AD patients carrying ApoE4 allele as compared with healthy adults. Elevated exosomal ApoE4 demonstrated significant inverse correlation with serum level of thyroid hormones and cognitive function. We analysed effects of ApoE4-containing peripheral exosomes on neural cells and neurological outputs in aged or thyroidectomised young mice. Ageing-associated hypothyroidism as well as acute thyroidectomy augmented transport of liver-derived ApoE4 reach exosomes into the brain, where ApoE4 activated nucleotide-binding oligomerisation domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome by increasing cholesterol level in neural cells. This, in turn, affected cognition, locomotion and mood. Our study reveals pathological potential of exosomes-mediated relocation of ApoE4 from the periphery to the brain, this process can represent potential therapeutic target.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Exossomos , Idoso , Envelhecimento , Doença de Alzheimer/metabolismo , Animais , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Exossomos/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Glândula Tireoide/metabolismo
6.
Commun Biol ; 5(1): 105, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115638

RESUMO

Stroke causes degeneration and death of neurones leading to the loss of motor function and frequent occurrence of cognitive impairment and depression. Lithium (Li+), the archetypal mood stabiliser, is neuroprotective in animal models of stroke, albeit underlying mechanisms remain unknown. We discover that Li+ inhibits activation of nucleotide-binding oligomerisation domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasomes in the middle cerebral artery occlusion (MCAO) stroke model in mice. This action of Li+ is mediated by two signalling pathways of AKT/GSK3ß/ß-catenin and AKT/FoxO3a/ß-catenin which converge in suppressing the production of reactive oxygen species (ROS). Using immunocytochemstry, MRI imaging, and cell sorting with subsequent mRNA and protein quantification, we demonstrate that Li+ decreases the infarct volume, improves motor function, and alleviates associated cognitive and depressive impairments. In conclusion, this study reveals molecular mechanisms of Li+ neuroprotection during brain ischaemia, thus providing the theoretical background to extend clinical applications of Li+ for treatment of ischemic stroke.


Assuntos
AVC Isquêmico/tratamento farmacológico , Lítio/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Disfunção Cognitiva/tratamento farmacológico , Depressão/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , AVC Isquêmico/patologia , Masculino , Camundongos , Camundongos Transgênicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Aleatória
7.
Commun Biol ; 4(1): 525, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33953326

RESUMO

Iron is the fundamental element for numerous physiological functions. Plasmalemmal divalent metal ion transporter 1 (DMT1) is responsible for cellular uptake of ferrous (Fe2+), whereas transferrin receptors (TFR) carry transferrin (TF)-bound ferric (Fe3+). In this study we performed detailed analysis of the action of Fe ions on cytoplasmic free calcium ion concentration ([Ca2+]i) in astrocytes. Administration of Fe2+ or Fe3+ in µM concentrations evoked [Ca2+]i in astrocytes in vitro and in vivo. Iron ions trigger increase in [Ca2+]i through two distinct molecular cascades. Uptake of Fe2+ by DMT1 inhibits astroglial Na+-K+-ATPase, which leads to elevation in cytoplasmic Na+ concentration, thus reversing Na+/Ca2+ exchanger and thereby generating Ca2+ influx. Uptake of Fe3+ by TF-TFR stimulates phospholipase C to produce inositol 1,4,5-trisphosphate (InsP3), thus triggering InsP3 receptor-mediated Ca2+ release from endoplasmic reticulum. In summary, these findings reveal the mechanisms of iron-induced astrocytic signalling operational in conditions of iron overload.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Compostos Férricos/farmacologia , Compostos Ferrosos/farmacologia , Receptores da Transferrina/metabolismo , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Transporte Biológico , Camundongos , Camundongos Endogâmicos C57BL , Sódio/metabolismo
8.
Brain Res ; 1752: 147234, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33412145

RESUMO

With the industrial development and progressive increase in environmental pollution, the mankind overexposure to heavy metals emerges as a pressing public health issue. Excessive intake of heavy metals, such as arsenic (As), manganese (Mn), mercury (Hg), aluminium (Al), lead (Pb), nickel (Ni), bismuth (Bi), cadmium (Cd), copper (Cu), zinc (Zn), and iron (Fe), is neurotoxic and it promotes neurodegeneration. Astrocytes are primary homeostatic cells in the central nervous system. They protect neurons against all types of insults, in particular by accumulating heavy metals. However, this makes astrocytes the main target for heavy metals neurotoxicity. Intake of heavy metals affects astroglial homeostatic and neuroprotective cascades including glutamate/GABA-glutamine shuttle, antioxidative machinery and energy metabolism. Deficits in these astroglial pathways facilitate or even instigate neurodegeneration. In this review, we provide a concise outlook on heavy metal-induced astrogliopathies and their association with major neurodegenerative disorders. In particular, we focus on astroglial mechanisms of iron-induced neurotoxicity. Iron deposits in the brain are detected in main neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Accumulation of iron in the brain is associated with motor and cognitive impairments and iron-induced histopathological manifestations may be considered as the potential diagnostic biomarker of neurodegenerative diseases. Effective management of heavy metal neurotoxicity can be regarded as a potential strategy to prevent or retard neurodegenerative pathologies.


Assuntos
Astrócitos/metabolismo , Astrócitos/patologia , Metais Pesados/toxicidade , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Exposição Ambiental/efeitos adversos , Humanos
9.
Function (Oxf) ; 2(2): zqab003, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35330817

RESUMO

Metal implants are used worldwide, with millions of nails, plates, and fixtures grafted during orthopedic surgeries. Iron is the most common element of these metal implants. As time passes, implants can be corroded and iron can be released. Ionized iron permeates the surrounding tissues and enters circulation; importantly, iron ions pass through the blood-brain barrier. Can iron from implants represent a risk factor for neurological diseases? This remains an unanswered question. In this study, we discovered that patients with metal implants delivered through orthopedic surgeries have higher incidence of Parkinson's disease or ischemic stroke compared to patients who underwent similar surgeries but did not have implants. Concentration of serum iron and ferritin was increased in subjects with metal implants. In experiments in vivo, we found that injection of iron dextran selectively decreased the presence of divalent metal transporter 1 (DMT1) in neurons through increasing the expression of Ndfip1, which degrades DMT1 and does not exist in glial cells. At the same time, excess of iron increased expression of DMT1 in astrocytes and microglial cells and triggered reactive astrogliosis and microgliosis. Facing the attack of excess iron, glial cells act as neuroprotectors to accumulate more extracellular iron by upregulating DMT1, whereas neurons limit iron uptake through increasing DMT1 degradation. Cerebral accumulation of iron in animals is associated with impaired cognition, locomotion, and mood. Excess iron from surgical implants thus can affect neural cells and may be regarded as a risk factor for neurodegeneration.


Assuntos
Ferro , Neurônios , Animais , Ferro/metabolismo , Neurônios/metabolismo , Neuroglia/metabolismo , Barreira Hematoencefálica/metabolismo , Doença Iatrogênica
10.
Sci Rep ; 10(1): 16073, 2020 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-32999319

RESUMO

Sexual dimorphism is evident in brain structure, size, and function throughout multiple species. Here, we tested whether cerebrospinal fluid entry into the glymphatic system, a network of perivascular fluid transport that clears metabolic waste from the brain, was altered between male and female mice. We analyze glymphatic influx in 244 young reproductive age (2-4 months) C57BL/6 mice. We found no male/female differences in total influx under anesthesia, or across the anterior/posterior axis of the brain. Circadian-dependent changes in glymphatic influx under ketamine/xylazine anesthesia were not altered by sex. This was not true for diurnal rhythms under pentobarbital and avertin, but both still showed daily oscillations independent of biological sex. Finally, although glymphatic influx decreases with age there was no sex difference in total influx or subregion-dependent tracer distribution in 17 middle aged (9-10 months) and 36 old (22-24 months) mice. Overall, in healthy adult C57BL/6 mice we could not detect male/female differences in glymphatic influx. This finding contrasts the gender differences in common neurodegenerative diseases. We propose that additional sex-dependent co-morbidities, such as chronic stress, protein misfolding, traumatic brain injury or other pathological mechanisms may explain the increased risk for developing proteinopathies rather than pre-existing suppression of glymphatic influx.


Assuntos
Sistema Glinfático/fisiologia , Envelhecimento/fisiologia , Anestesia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/fisiologia , Líquido Cefalorraquidiano/fisiologia , Ritmo Circadiano/fisiologia , Feminino , Sistema Glinfático/diagnóstico por imagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Caracteres Sexuais
12.
Neurosci Bull ; 36(11): 1259-1270, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32506374

RESUMO

Chronic loss of sleep damages health and disturbs the quality of life. Long-lasting sleep deprivation (SD) as well as sleep abnormalities are substantial risk factors for major depressive disorder, although the underlying mechanisms are not clear. Here, we showed that chronic SD in mice promotes a gradual elevation of extracellular ATP, which activates astroglial P2X7 receptors (P2X7Rs). Activated P2X7Rs, in turn, selectively down-regulated the expression of 5-HT2B receptors (5-HT2BRs) in astrocytes. Stimulation of P2X7Rs induced by SD selectively suppressed the phosphorylation of AKT and FoxO3a in astrocytes, but not in neurons. The over-expression of FoxO3a in astrocytes inhibited the expression of 5-HT2BRs. Down-regulation of 5-HT2BsRs instigated by SD suppressed the activation of STAT3 and relieved the inhibition of Ca2+-dependent phospholipase A2. This latter cascade promoted the release of arachidonic acid and prostaglandin E2. The depression-like behaviors induced by SD were alleviated in P2X7R-KO mice. Our study reveals the mechanism underlying chronic SD-induced depression-like behaviors and suggests 5-HT2BRs as a key target for exploring therapeutic strategies aimed at the depression evoked by sleep disorders.


Assuntos
Astrócitos , Depressão/fisiopatologia , Receptor 5-HT2B de Serotonina/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Privação do Sono , Trifosfato de Adenosina , Animais , Astrócitos/citologia , Proteína Forkhead Box O3/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica v-akt/metabolismo , Qualidade de Vida , Fator de Transcrição STAT3/metabolismo , Serotonina
13.
Neurochem Int ; 134: 104689, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31968217

RESUMO

The alcoholism and major depressive disorder are common comorbidity, with alcohol-induced depressive symptoms being eased by selective serotonin re-uptake inhibitors (SSRIs), although the mechanisms underlying pathology and therapy are poorly understood. Chronic alcohol consumption affects the activity of serotonin 2C receptors (5-HT2CR) by regulating adenosine deaminases acting on RNA (ADARs) in neurons. Astrogliopathic changes contribute to alcohol addiction, while decreased release of ATP from astrocytes can trigger depressive-like behaviours in mice. In this study, we discovered that chronic alcohol treatment increased editing of RNA of 5-HT2CR via up-regulating the expression of ADAR2, consequently reducing the release of ATP from astrocytes induced by 5-HT2CR agonist, MK212. Moreover, SSRI antidepressant fluoxetine decreased the expression of ADAR2 through the transactivation of EGFR/PI3K/AKT/cFos signalling pathway. The increased release of astroglial ATP by MK212 which was suppressed by chronic alcohol consumption, and reduction in ADAR2 activity eliminated the RNA editing of 5-HT2CR increased by alcohol in vitro and recovered the release of ATP from astrocytes induced by MK212. Meanwhile, fluoxetine improved the behavioural and motor symptoms induced by alcohol addiction and decreased the alcohol intake. Our study suggests that the astrocytic 5-HT2CR contribute to alcohol addiction; fluoxetine thus can be used to alleviate depression, treat alcohol addiction and improve motor coordination.


Assuntos
Álcoois/farmacologia , Comportamento Animal/efeitos dos fármacos , Fluoxetina/farmacologia , Receptor 5-HT2C de Serotonina/efeitos dos fármacos , Alcoolismo/tratamento farmacológico , Alcoolismo/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Transtorno Depressivo Maior/tratamento farmacológico , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Edição de RNA/efeitos dos fármacos , Receptor 5-HT2C de Serotonina/metabolismo , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos
14.
Neurochem Int ; 129: 104500, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31295509

RESUMO

Spinal cord injury (SCI) is a serious central nervous system (CNS) trauma that results in permanent and severe disability. The extracellular matrix (ECM) can affect the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) by interacting with the ERK integrin subunits. In this study, we built a model of SCI with glial fibrillary acidic protein-green fluorescent protein (GFAP-GFP) and thymus cell antigen 1-yellow fluorescent protein-H (Thy1-YFPH) in mice that express specific transgenes in their astrocytes or neurons. Then, we collected spinal cord neurons or astrocytes by fluorescence-activated cell sorting (FACS). In this way, we investigated the SCI-induced phosphorylation of ERK1/2 and epidermal growth factor receptor (EGFR) in neurons and astrocytes, and we discovered that the SCI-induced EGFR signaling pathways differed between neurons and astrocytes. In the present study, we found that the Src-dependent phosphorylation of EGFR induced by SCI occurred only in neurons, not in astrocytes. This phenomenon may be due to the involvement of Thy-1, which promoted the binding between Src and EGFR in neurons after SCI. In addition, the expression of the integrin subunits after SCI differed between neurons and astrocytes. Our present study shows that the EGFR signaling pathway triggered by SCI in neurons differed from the EGFR signaling pathway triggered in astrocytes, a finding that may help to pave the way for clinical trials of therapies that inhibit EGFR signaling pathways after SCI.


Assuntos
Astrócitos/metabolismo , Receptores ErbB/fisiologia , Matriz Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neurônios/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Regulação da Expressão Gênica , Integrinas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas do Tecido Nervoso/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional , Subunidades Proteicas , Receptores de Vitronectina/antagonistas & inibidores , Traumatismos da Medula Espinal/genética
15.
Cell Calcium ; 76: 122-128, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30469142

RESUMO

Spinal cord injury (SCI) causes long-term disability and has no effective clinical treatment. After SCI, extracellular adenosine triphosphate (ATP) leads to an influx of extracellular Ca2+, and this Ca2+ overload causes neuronal toxicosis and apoptosis. The biological functions of leptin have been widely investigated in the central nervous system. In this study, we discovered that the administration of leptin could improve locomotor recovery following SCI. The aim of this study was to determine the neuroprotective mechanism of leptin in vivo and in vitro. The neuronal apoptosis and Ca2+ imaging signal induced by ATP were suppressed by leptin, due to elevated caveolin-1 expression. In vivo two-photon observations revealed that leptin reduced the neuronal Ca2+ imaging signal in the exposed spinal cords of live Thy1-YFP mice. In conclusion, leptin promotes locomotor functional recovery and suppresses neuronal impairment after SCI, suggesting that leptin has a promising clinical therapeutic value for treatment of SCI.


Assuntos
Caveolina 1/biossíntese , Leptina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Substâncias Protetoras/farmacologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Cálcio/análise , Cálcio/metabolismo , Caveolina 1/genética , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Injeções Intraperitoneais , Leptina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Substâncias Protetoras/administração & dosagem , Relação Estrutura-Atividade
16.
Neurochem Res ; 43(4): 848-856, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29404840

RESUMO

Spinal cord injury (SCI) is a type of long-term disability with a high morbidity rate in clinical settings for which there is no effective clinical treatment to date. Usually, lithium is used as a popular mood stabilizer. Recently, growing evidence has shown that lithium has clear neuroprotective effects after SCI, and the administration of lithium can effectively improve locomotor recovery. However, the exact neuroprotective mechanism of lithium is still not understood. Glycogen synthase kinase-3 beta (GSK3ß) is a serine/threonine kinase that plays an important role in the neuroprotective effects of lithium both in vivo and in vitro. In this study, we discovered that lithium inhibits GSK3ß activity through two different signaling pathways in spinal cord neurons. In the acute phase, lithium inhibited GSK3ß activity by stimulating phosphorylation of AKT; in the chronic phase, we first discovered that lithium additionally upregulated the expression of Na+, K+-ATPase α1 (NKA α1), which had an inhibitory effect on GSK3ß activity by inducing the expression of glucocorticoid inducible kinase 1 (SGK1). SGK1 is well known as a regulator of the GSK3ß/ß-catenin signaling pathway. Moreover, the suppressed activity of GSK3ß increased the level of ß-catenin in the cytoplasm, which gave rise to the translocation of the freely stabilized ß-catenin to the nucleus. In addition, the accumulation of ß-catenin in the nucleus had the benefits of neuronal survival. Hopefully our findings from this study are beneficial in revealing the neuroprotective mechanism of lithium and in offering novel targets for the development of new SCI therapeutic drugs.


Assuntos
Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Cloreto de Lítio/farmacologia , Cloreto de Lítio/uso terapêutico , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Glicogênio Sintase Quinase 3 beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/enzimologia , Transdução de Sinais/fisiologia , Traumatismos da Medula Espinal/enzimologia
17.
RSC Adv ; 8(36): 20095-20107, 2018 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35541692

RESUMO

Hydrothermal carbon nanospheres were prepared by introducing opal into the hydrothermal carbonization system of hydroxypropyl methyl cellulose (HPMC). Then the effects of opal on hydrothermal carbonization of HPMC were investigated after different reaction durations (105-240 min). The reaction products were characterized by elemental analysis, gas chromatography-mass spectrometry (GC-MS), scanning electron microscopy (SEM), energy dispersive X-ray spectroscopy (EDS), Fourier transform infrared spectroscopy (FT-IR) and N2 adsorption-desorption. Results of elemental analysis indicated that the H (hydrogen) and O (oxygen) content of HPMC decreased through dehydration, demethylation, decarbonylation and hydrolysis reactions, forming hydrochar with higher carbon content. The addition of opal was confirmed to accelerate the hydrolysis of HPMC. N2 adsorption-desorption tests and SEM analysis showed that opal with a large specific surface area adsorbed HPMC hydrolysis products, such as furans, and facilitated furan cyclodehydration on its surfaces to form cross-linked carbons, which contributed to the quick formation of hydrochar. Moreover, the adsorption by opal also inhibited hydrochar aggregation, so the final hydrothermal carbon spheres had sizes of 20-100 nm.

18.
Front Psychiatry ; 9: 734, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30666218

RESUMO

The long-lasting loss of sleep is a generally acknowledged risk factor for the occurrence of major depressive disorder (MDD), whereas sleep abnormalities being a key clinic symptom of the MDD. In our previous work, we demonstrated that the sleep deprivation (SD) stimulates activation of nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasomes as well as the release of IL-1ß and IL-18 from astrocytes. However, the underlying mechanism connecting SD and MDD still requires further study. Apart of the secretion of the pro-inflammatory cytokines, SD affects production of brain-derived neurotrophic factor (BDNF) while release of BDNF from astrocytes appears a key contributor to mood disorders. If and how the activation of NLRP3 inflammasome following SD affects the level of BDNF remains unknown. Antidepressant fluoxetine acts through astroglial 5-hydroxytryptamine receptor 2B (5-HT2B); these receptors are also related to the sleep-wake cycle. Contribution of leptin to MDD has been discovered recently, although the mechanistic links between leptin and the depressive-like behaviors has not been revealed. In this study, we discovered: (i) that activation of NLRP3 inflammasome was involved in the depressive-like behaviors induced by SD; (ii) decrease in BDNF following SD required the activation of NLRP3 inflammasomes; (iii) leptin augmented the anti-depressive action of fluoxetine through an increase in expression of astrocytic 5-HT2B receptors. We suggest that decrease in BDNF by the activated NLRP3 inflammasomes in astrocytes is the key pathological event of the depressive-like behaviors induced by SD, while the combined treatment with fluoxetine and leptin improves therapeutic outcome for the depression induced by SD.

19.
J Neurochem ; 2017 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-29222907

RESUMO

It is well known that sleep disorders are harmful to people's health and performance, and growing evidence suggests that sleep deprivation (SD) can trigger neuroinflammation in the brain. The nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasome is reported to be relevant to the neuroinflammation induced by SD, but the regulatory signaling that governs the NLRP3 inflammasome in SD is still unknown. Meanwhile, whether the regulatory action of antidepressants in astrocytes could affect the neuroinflammation induced by SD also remains obscure. In this study, we were the first to discover that the antidepressant fluoxetine, a type of specific serotonin reuptake inhibitor widely used in clinical practice, could suppress the neuroinflammation and neuronal apoptosis induced by SD. The main findings from this study are as follows: (i) SD stimulated the expression of activated NLRP3 inflammasomes and the maturation of IL-1ß/18 via suppressing the phosphorylation of STAT3 in astrocytes; (ii) SD decreased the activation of AKT and stimulated the phosphorylation of GSK-3ß, which inhibited the phosphorylation of STAT3; (iii) the NLRP3 inflammasome expression stimulated by SD was partly mediated by the P2X7 receptor; (iv) an agonist of STAT3 could significantly abolish the expression of NLRP3 inflammasomes induced by an agonist of the P2X7 receptor in primary cultured astrocytes; (v) the administration of fluoxetine could reverse the stimulation of NLRP3 inflammasome expression and function by SD through elevating the activation of STAT3. In conclusion, our present research suggests the promising possibility that fluoxetine could ameliorate the neuronal impairment induced by SD.

20.
ACS Appl Mater Interfaces ; 9(30): 25291-25297, 2017 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-28695728

RESUMO

As an electrocatalyst, conventional 2H-phase MoS2 suffers from limited active sites and inherently low electroconductivity. Phase transitions from 2H to 1T have been proposed as an effective strategy for optimization of the catalytic activity. However, complicated chemical exfoliation is generally involved. Here, MoS2 heterogeneous-phase nanosheets with a 1T phase (1T/2H-MoS2) generated in situ were prepared through a facile hydrothermal method. The locally introduced 1T-phase MoS2 can not only contribute more active sites but also markedly promote the electronic conductivity. Because of this unique structure, the as-synthesized 1T/2H-MoS2 nanosheets exhibit remarkable performance for the hydrogen evolution reaction with a small overpotential of 220 mV at 10 mA/cm2, a small Tafel slope of 61 mV/decade, and robust stability. This work facilitates the development of a two-dimensional heterogeneous nanostructure with enhanced applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA