Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Mol Psychiatry ; 28(7): 3044-3054, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36991130

RESUMEN

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.


Asunto(s)
Trastorno Bipolar , Manía , Humanos , Animales , Ratones , Modelos Animales de Enfermedad , Ácido Valproico , Privación de Sueño
2.
Neurochem Res ; 43(4): 848-856, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29404840

RESUMEN

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.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Cloruro de Litio/farmacología , Cloruro de Litio/uso terapéutico , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/enzimología , Transducción de Señal/fisiología , Traumatismos de la Médula Espinal/enzimología
3.
J Neurochem ; 2017 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-29222907

RESUMEN

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.

4.
J Neurosci Res ; 94(10): 924-35, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27316329

RESUMEN

Spinal cord injury (SCI) causes long-term disability and has no clinically effective treatment. After SCI, adenosine triphosphate (ATP) may be released from neuronal cells and astrocytes in large amounts. Our previous studies have shown that the extracellular release of ATP increases the phosphorylation of cytosolic phospholipase A2 (cPLA2 ) and triggers the rapid release of arachidonic acid (AA) and prostaglandin E2 (PGE2) via the stimulation of epidermal growth factor receptor (EGFR) and the downstream phosphorylation of extracellular-regulated protein kinases 1 and 2. Leptin, a glycoprotein, induces the activation of the Janus kinase (JAK2)/signal transducers and activators of transcription-3 (Stat3) pathway via the leptin receptor. In this study, we found that 1) prolonged leptin treatment suppressed the ATP-stimulated release of AA and PGE2 from cultured spinal cord astrocytes; 2) leptin elevated the expression of caveolin-1 (Cav-1) via the JAK2/Stat3 signaling pathway; 3) Cav-1 blocked the interaction between Src and EGFR, thereby inhibiting the phosphorylation of EGFR and cPLA2 and attenuating the release of AA or PGE2; 4) pretreatment with leptin decreased ;he level of apoptosis and the release of interleukin-6 from cocultured neurons and astrocytes; and 5) leptin improved the recovery of locomotion in mice after SCI. Our results highlight leptin as a promising therapeutic agent for SCI. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Adenosina Trifosfato/farmacología , Astrocitos/efectos de los fármacos , Leptina/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Médula Espinal/citología , Animales , Proteínas de Unión al Calcio/metabolismo , Caveolina 1/genética , Caveolina 1/metabolismo , Células Cultivadas , Cultura , Modelos Animales de Enfermedad , Receptores ErbB/genética , Receptores ErbB/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
Ann Neurol ; 76(6): 845-61, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25204284

RESUMEN

OBJECTIVE: In the brain, protein waste removal is partly performed by paravascular pathways that facilitate convective exchange of water and soluble contents between cerebrospinal fluid (CSF) and interstitial fluid (ISF). Several lines of evidence suggest that bulk flow drainage via the glymphatic system is driven by cerebrovascular pulsation, and is dependent on astroglial water channels that line paravascular CSF pathways. The objective of this study was to evaluate whether the efficiency of CSF-ISF exchange and interstitial solute clearance is impaired in the aging brain. METHODS: CSF-ISF exchange was evaluated by in vivo and ex vivo fluorescence microscopy and interstitial solute clearance was evaluated by radiotracer clearance assays in young (2-3 months), middle-aged (10-12 months), and old (18-20 months) wild-type mice. The relationship between age-related changes in the expression of the astrocytic water channel aquaporin-4 (AQP4) and changes in glymphatic pathway function was evaluated by immunofluorescence. RESULTS: Advancing age was associated with a dramatic decline in the efficiency of exchange between the subarachnoid CSF and the brain parenchyma. Relative to the young, clearance of intraparenchymally injected amyloid-ß was impaired by 40% in the old mice. A 27% reduction in the vessel wall pulsatility of intracortical arterioles and widespread loss of perivascular AQP4 polarization along the penetrating arteries accompanied the decline in CSF-ISF exchange. INTERPRETATION: We propose that impaired glymphatic clearance contributes to cognitive decline among the elderly and may represent a novel therapeutic target for the treatment of neurodegenerative diseases associated with accumulation of misfolded protein aggregates.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Circulación Cerebrovascular/fisiología , Tasa de Depuración Metabólica/fisiología , Envejecimiento/patología , Animales , Acuaporina 4/metabolismo , Encéfalo/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neuroglía/metabolismo , Neuroglía/patología
6.
J Neurosci Res ; 92(8): 1078-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24687862

RESUMEN

After spinal cord injury (SCI), the formation of glial scar is a complex process that is attributed primarily to astrocytic proliferation, but the mechanism of astrocytes proliferation is still unclear. Fibronectin is a large extracellular glycoprotein that helps organize the matrix protein, and its main membrane receptor is the α5 ß1 integrin subunit. In this study, fibronectin stimulated spinal cord astrocytic proliferation from two directions: fibronectin increased astrocytic proliferation via α5 ß1 integrin receptor, and fibronectin upregulated the expression of P2Y1 receptor, and adenosine triphosphate (ATP) could enhance the astrocytic proliferation and induce more release of arachidonic acid and prostaglandin E2 via P2Y1. The upregulation of P2Y1 by fibronectin required [Ca2+]i and the activation of integrin link kinase (ILK) and Akt. We found that [Ca2+]i stimulated by fibronectin was α5 ß1 integrin receptor dependent and that the phosphorylation of Akt or extracellular signal-regulated protein kinase (ERK1/2) induced by fibronectin mediated the activation of cAMP response element-binding protein (CREB) and signal transducer and activator of transcription 3 (Stat3). Our research suggests that the release of fibronectin and ATP could stimulate the spinal cord astrocytic proliferation after SCI, and the expression of P2Y1 increased by fibronectin would provide more sites for ATP, which could aggravate the proliferation and inflammation of spinal cord astrocytes.


Asunto(s)
Astrocitos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fibronectinas/farmacología , Receptores Purinérgicos P2Y1/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Astrocitos/citología , Astrocitos/metabolismo , Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Integrina alfa5beta1/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Regulación hacia Arriba/efectos de los fármacos
7.
Signal Transduct Target Ther ; 9(1): 30, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38331979

RESUMEN

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.


Asunto(s)
Trastorno Depresivo Mayor , Humanos , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/prevención & control , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal
8.
Neural Regen Res ; 19(5): 1126-1133, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37862218

RESUMEN

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.

9.
J Psychiatr Res ; 161: 188-198, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36933445

RESUMEN

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.


Asunto(s)
Trastornos por Estrés Postraumático , Ratones , Masculino , Animales , Trastornos por Estrés Postraumático/metabolismo , Leptina , Ratones Transgénicos , Especies Reactivas de Oxígeno , Glucógeno Sintasa Quinasa 3 beta
10.
Commun Biol ; 5(1): 105, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35115638

RESUMEN

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.


Asunto(s)
Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Litio/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Disfunción Cognitiva/tratamiento farmacológico , Depresión/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Accidente Cerebrovascular Isquémico/patología , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Aleatoria
11.
Cell Death Dis ; 13(4): 406, 2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35468877

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Exosomas , Anciano , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Animales , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Exosomas/metabolismo , Humanos , Hígado/metabolismo , Ratones , Glándula Tiroides/metabolismo
12.
Glia ; 59(4): 664-74, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21294165

RESUMEN

Traumatic spinal cord injury is characterized by an immediate, irreversible loss of tissue at the lesion site, as well as a secondary expansion of tissue damage over time. Although secondary injury should, in principle, be preventable, no effective treatment options currently exist for patients with acute spinal cord injury (SCI). Excessive release of ATP by the traumatized tissue, triggers the rapid release of arachidonic acid (AA) and prostaglandin E2 (PGE2), and has beenimplicated in acute and chronic neuropathic pain and inflammation. But the intracellular pathways between ATP and PGE2 remain largely unknown. We have explored the signaling events involved in this synthesis by primarily culturing spinal cord astrocytes: (1) we determined significant PGE2 production increased by ATP is mainly via Subtype 1 of P2 purinoceptors (P2Y1) but not P2Y2; (2) we found that ATP strongly increased the level of intracellular Ca(2+) via P2Y1 receptor; (3) we indicated that ATP stimulates the definitely release of AA and PGE2 which involved the transactivation of epidermal growth factor (EGF) receptor, the phosphorylation of extracellular-regulated protein kinases 1 and 2 (ERK(1/2) ) and the activation of cytosolic phospholipase A(2) (cPLA(2) ); (4) we examined ATP could increase the phosphorylation of Akt via P2Y1 receptor which also depend on the transactivation of EGFR, but the activation of Akt has no effect on the downstream of cPLA(2) phosphorylation. ATP induced by SCI could mobilize the release of AA and PGE2. And inhibition of PGE2 release reduces behavioral signs of pain after SCI and peripheral nerve injury.


Asunto(s)
Adenosina Trifosfato/farmacología , Astrocitos/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Adenosina Trifosfato/metabolismo , Análisis de Varianza , Animales , Astrocitos/efectos de los fármacos , Western Blotting , Técnicas para Inmunoenzimas , Inmunohistoquímica , Inmunoprecipitación , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos
13.
Brain Res ; 1752: 147234, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33412145

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Metales Pesados/toxicidad , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Exposición a Riesgos Ambientales/efectos adversos , Humanos
14.
Function (Oxf) ; 2(2): zqab003, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35330817

RESUMEN

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.


Asunto(s)
Hierro , Neuronas , Animales , Hierro/metabolismo , Neuronas/metabolismo , Neuroglía/metabolismo , Barrera Hematoencefálica/metabolismo , Enfermedad Iatrogénica
15.
Commun Biol ; 4(1): 525, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953326

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Compuestos Férricos/farmacología , Compuestos Ferrosos/farmacología , Receptores de Transferrina/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Transporte Biológico , Ratones , Ratones Endogámicos C57BL , Sodio/metabolismo
16.
Neurochem Int ; 134: 104689, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31968217

RESUMEN

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.


Asunto(s)
Alcoholes/farmacología , Conducta Animal/efectos de los fármacos , Fluoxetina/farmacología , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Alcoholismo/tratamiento farmacológico , Alcoholismo/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Edición de ARN/efectos de los fármacos , Receptor de Serotonina 5-HT2C/metabolismo , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos
17.
Sci Rep ; 10(1): 16073, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32999319

RESUMEN

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.


Asunto(s)
Sistema Glinfático/fisiología , Envejecimiento/fisiología , Anestesia , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Líquido Cefalorraquídeo/fisiología , Ritmo Circadiano/fisiología , Femenino , Sistema Glinfático/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Caracteres Sexuales
18.
Neurosci Bull ; 36(11): 1259-1270, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32506374

RESUMEN

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.


Asunto(s)
Astrocitos , Depresión/fisiopatología , Receptor de Serotonina 5-HT2B/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Privación de Sueño , Adenosina Trifosfato , Animales , Astrocitos/citología , Proteína Forkhead Box O3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Oncogénica v-akt/metabolismo , Calidad de Vida , Factor de Transcripción STAT3/metabolismo , Serotonina
19.
Neurochem Int ; 129: 104500, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31295509

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Receptores ErbB/fisiología , Matriz Extracelular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neuronas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Regulación de la Expresión Génica , Integrinas/fisiología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Ratones , Ratones Endogámicos , Proteínas del Tejido Nervioso/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Subunidades de Proteína , Receptores de Vitronectina/antagonistas & inhibidores , Traumatismos de la Médula Espinal/genética
20.
Connect Tissue Res ; 49(6): 464-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19085247

RESUMEN

Integrins are a class of cell adhesion molecules that regulate interactions between cells and their extracellular matrix (ECM). Several specific integrin receptors have been identified in intervertebral discs, including the fibronectin-binding integrin receptors alpha(5) beta(1), alpha(v) beta(3) and the collagen-binding integrin receptors alpha(1) beta(1), alpha(2) beta(1), and, alpha(v) beta(1). But the integrins expressions in degenerated intervertebral discs are still unknown. In our study, the expressions of alpha(1), alpha(2), alpha(5), alpha(v), beta(1), beta(3) integrin subunits, collagens, and fibronectin in normal and herniated intervertebral discs of human were determined. Specimens of human lumbar intervertebral discs were divided into 3 groups: normal discs (n = 10), protrusion (n = 15), and extrusion (n = 15). Real-time quantitative reverse transcription polymerase chain reaction (RT-PCR) and immunoprecipitation were used to evaluate the alpha(1), alpha(2), alpha(5),alpha(v), beta(1), and beta(3) integrin subunits messenger ribonucleic acid (mRNA) and protein expressions. RT-PCR was also performed to measure the mRNA level of collagen I, collagen II, and fibronectin. The expressions of alpha(5) and beta(1) subunits were increased in herniated discs, especially in the discs of extrusion. But as to alpha(1), alpha(2), alpha(v) and beta(3), their expressions had no difference among the discs. Fibronectin, whose binding integrin receptor was alpha(5) beta(1) was also increased. And in herniated discs, the collagen I was increased, but the collagen II was decreased. The expressions of some integrin subunits were increased in herniated discs. These results may be attributed to the interaction between cells and the ECM in the process of degeneration.


Asunto(s)
Integrinas/biosíntesis , Desplazamiento del Disco Intervertebral/metabolismo , Disco Intervertebral/metabolismo , Adulto , Colágeno/metabolismo , Fibronectinas/metabolismo , Humanos , Persona de Mediana Edad , Subunidades de Proteína/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA