Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ACS Chem Neurosci ; 15(1): 31-55, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38118278

RESUMEN

Neuroinflammation is a key pathophysiological feature of stroke-associated brain injury. A local innate immune response triggers neuroinflammation following a stroke via activating inflammasomes. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has been heavily implicated in stroke pathobiology. Following a stroke, several stimuli have been suggested to trigger the assembly of the NLRP3 inflammasome. Recent studies have advanced the understanding and revealed several new players regulating NLRP3 inflammasome-mediated neuroinflammation. This article discussed recent advancements in NLRP3 assembly and highlighted stroke-induced mitochondrial dysfunction as a major checkpoint to regulating NLRP3 activation. The NLRP3 inflammasome activation leads to caspase-1-dependent maturation and release of IL-1ß, IL-18, and gasdermin D. In addition, genetic or pharmacological inhibition of the NLRP3 inflammasome activation and downstream signaling has been shown to attenuate brain infarction and improve the neurological outcome in experimental models of stroke. Several drug-like small molecules targeting the NLRP3 inflammasome are in different phases of development as novel therapeutics for various inflammatory conditions, including stroke. Understanding how these molecules interfere with NLRP3 inflammasome assembly is paramount for their better optimization and/or development of newer NLRP3 inhibitors. In this review, we summarized the assembly of the NLRP3 inflammasome and discussed the recent advances in understanding the upstream regulators of NLRP3 inflammasome-mediated neuroinflammation following stroke. Additionally, we critically examined the role of the NLRP3 inflammasome-mediated signaling in stroke pathophysiology and the development of therapeutic modalities to target the NLRP3 inflammasome-related signaling for stroke treatment.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Neuroinflamatorias , Accidente Cerebrovascular/tratamiento farmacológico , Interleucina-1beta/metabolismo
2.
Life Sci ; 316: 121370, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36640902

RESUMEN

AIM: The combined impact of traditional and non-traditional risk factors of type 2 diabetes (T2D) on the development and progression of insulin resistance and associated complications is poorly understood. Therefore, we assessed the effect of moderately rich sugar diet coupled with environmental chemical exposure on the development and progression of T2D using Drosophila as a model organism. MAIN METHODS: We reared newly eclosed Drosophila males on a diet containing atrazine (20 µg/ml; non-traditional risk factor) and/or moderately high sucrose (0.5 M/1 M; to mimic binge eating, Traditional risk factor) for 20-30 days. Subsequently, we assessed diabetic parameters, oxidative stress parameters and also the abundance of advanced glycation end products (AGEs) along with their receptor (RAGE) in these flies. For diabetic cardiomyopathy, we examined the pericardin (tissue fibrosis marker) level in Drosophila heart. KEY FINDINGS: Flies reared on 20 µg/ml atrazine alone showed T2D hallmarks at 30 days. In contrast, flies reared on 0.5 M sucrose+ 20 µg/ml atrazine showed insulin resistance characterized by hyperglycemia and increased Drosophila insulin-like peptides along with reduced insulin signaling at 20 days, similar to those reared on high sucrose diet. In addition, both groups had high levels of oxidative stress and showed starvation response (converting triglycerides into fatty acids). Alarmingly, flies fed with sucrose+atrazine for 20 and 30 days had elevated pericardin in heart tissues, indicating early onset of diabetic complications such as cardiomyopathy. SIGNIFICANCE: Lifestyle-chemical exposure synergistically impairs glucose metabolism, affects organisms' redox state and leads to the early onset of T2D and associated complications like cardiomyopathy.


Asunto(s)
Atrazina , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Masculino , Drosophila/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Atrazina/toxicidad , Insulina/metabolismo , Estrés Oxidativo , Dieta , Productos Finales de Glicación Avanzada/metabolismo , Sacarosa/farmacología
3.
Toxicol Sci ; 190(1): 79-98, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-35993674

RESUMEN

Arsenic is an environmental contaminant with potential neurotoxicity. We previously reported that arsenic promoted hippocampal neuronal apoptosis, inducing cognitive loss. Here, we correlated it with tau pathology. We observed that environmentally relevant arsenic exposure increased tau phosphorylation and the principal tau kinase, glycogen synthase kinase-3 beta (GSK3ß), in the female rat hippocampal neurons. We detected the same in primary hippocampal neurons. Because a regulated estrogen receptor (ER) level and inflammation contributed to normal hippocampal functions, we examined their levels following arsenic exposure. Our ER screening data revealed that arsenic down-regulated hippocampal neuronal ERα. We also detected an up-regulated hippocampal interleukin-1 (IL-1) and its receptor, IL-1R1. Further, co-treating arsenic with the ERα agonist, 4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT), or IL-1R antagonist (IL-1Ra) resulted in reduced GSK3ß and p-tau, indicating involvement of decreased ERα and increased IL-1/IL-1R1 in tau hyperphosphorylation. We then checked whether ERα and IL-1/IL-1R1 had linkage, and detected that although PPT reduced IL-1 and IL-1R1, the IL-1Ra restored ERα, suggesting their arsenic-induced interdependence. We finally correlated this pathway with apoptosis and cognition. We observed that PPT, IL-1Ra and the GSK3ß inhibitor, LiCl, reduced hippocampal neuronal cleaved caspase-3 and TUNEL+ve apoptotic count, and decreased the number of errors during learning and increased the saving memory for Y-Maze test and retention performance for Passive avoidance test in arsenic-treated rats. Thus, our study reveals a novel mechanism of arsenic-induced GSK3ß-dependent tau pathology via interdependent ERα and IL-1/IL-1R1 signaling. It also envisages the protective role of ERα agonist and IL-1 inhibitor against arsenic-induced neurotoxicity.


Asunto(s)
Arsénico , Disfunción Cognitiva , Animales , Femenino , Ratas , Apoptosis , Arsénico/toxicidad , Arsénico/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Receptor alfa de Estrógeno/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hipocampo/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1/metabolismo , Fosforilación , Proteínas tau/metabolismo
4.
Mol Neurobiol ; 59(5): 2729-2744, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35175559

RESUMEN

We earlier reported that arsenic induced hippocampal neuronal loss, causing cognitive dysfunctions in male rats. This neuronal damage mechanism involved an altered bone morphogenetic protein (BMP2)/Smad and brain-derived neurotrophic factor (BDNF)/TrkB signaling. Susceptibility to toxicants is often sex-dependent, and hence we studied the comparative effects of arsenic in adult male and female rats. We observed that a lower dose of arsenic reduced learning-memory ability, examined through passive avoidance and Y-maze tests, in male but not female rats. Again, male rats exhibited greater learning-memory loss at a higher dose of arsenic. Supporting this, arsenic-treated male rats demonstrated larger reduction in the hippocampal NeuN and %-surviving neurons, together with increased apoptosis and altered BMP2/Smad and BDNF/TrkB pathways compared to their female counterparts. Since the primary female hormone, estrogen (E2), regulates normal brain functions, we next probed whether endogenous E2 levels in females offered resistance against arsenic-induced neurotoxicity. We used ovariectomized (OVX) rat as the model for E2 deficiency. We primarily identified that OVX itself induced hippocampal neuronal damage and cognitive decline, involving an increased BMP2/Smad and reduced BDNF/TrkB. Further, these effects appeared greater in arsenic + OVX compared to arsenic + sham (ovary intact) or OVX rats alone. The OVX-induced adverse effects were significantly reduced by E2 treatment. Overall, our study suggests that adult males could be more susceptible than females to arsenic-induced neurotoxicity. It also indicates that endogenous E2 regulates hippocampal BMP and BDNF signaling and restrains arsenic-induced neuronal dysfunctions in females, which may be inhibited in E2-deficient conditions, such as menopause or ovarian failure.


Asunto(s)
Arsénico , Estrógenos/metabolismo , Síndromes de Neurotoxicidad , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cognición , Estradiol/farmacología , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Ovariectomía , Ratas
5.
Mol Neurobiol ; 58(3): 1196-1211, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33106949

RESUMEN

Thyroid hormone (TH) is essential for brain development, and hypothyroidism induces cognitive deficits in children and young adults. However, the participating mechanisms remain less explored. Here, we examined the molecular mechanism, hypothesizing the involvement of a deregulated autophagy and apoptosis pathway in hippocampal neurons that regulate cognitive functions. Therefore, we used a rat model of developmental hypothyroidism, generated through methimazole treatment from gestation until young adulthood. We detected that methimazole stimulated the autophagy mechanism, characterized by increased LC3B-II, Beclin-1, ATG7, and ATG5-12 conjugate and decreased p-mTOR/mTOR and p-ULK1/ULK1 autophagy regulators in the hippocampus of developing and young adult rats. This methimazole-induced hippocampal autophagy could be inhibited by thyroxine treatment. Subsequently, probing the upstream mediators of autophagy revealed an increased hippocampal neuroinflammation, marked by upregulated interleukin (IL)-1alpha and beta and activated microglial marker, Iba1, promoting neuronal IL-1 receptor-1 expression. Hence, IL-1R-antagonist (IL-1Ra), which reduced hippocampal neuronal IL-1R1, also inhibited the enhanced autophagy in hypothyroid rats. We then linked these events with hypothyroidism-induced apoptosis and loss of hippocampal neurons, where we observed that like thyroxine, IL-1Ra and autophagy inhibitor, 3-methyladenine, reduced the cleaved caspase-3 and TUNEL-stained apoptotic neurons and enhanced Nissl-stained neuronal count in methimazole-treated rats. We further related these molecular results with cognition through Y-maze and passive avoidance tests, demonstrating an IL-1Ra and 3-methyladenine-mediated improvement in learning-memory performances of the hypothyroid rats. Taken together, our study enlightens the critical role of neuroinflammation-dependent autophagy mechanism in TH-regulated hippocampal functions, disrupted in developmental hypothyroidism.


Asunto(s)
Apoptosis , Autofagia , Disfunción Cognitiva/etiología , Hipocampo/patología , Hipotiroidismo/complicaciones , Hipotiroidismo/patología , Interleucina-1/metabolismo , Neuronas/patología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Hipotiroidismo/sangre , Hipotiroidismo/fisiopatología , Inflamación/patología , Memoria/efectos de los fármacos , Metimazol/farmacología , Microglía/efectos de los fármacos , Microglía/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Ratas Wistar , Serina-Treonina Quinasas TOR/metabolismo , Tiroxina/sangre , Triyodotironina/sangre
6.
Biol Trace Elem Res ; 196(2): 654-661, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31925741

RESUMEN

Arsenic, mercury and lead are the environmental toxicants which exert their toxic effects through binding with certain proteins including their structures and functions. The toxicity of these heavy metal results is associated to its interaction with the metalloenzymes. They replace the essential metals required for normal biochemical functions of enzymes. The superoxide dismutase (SOD), a metalloenzyme, requires certain cofactors such as Cu2+ and Zn2+ for their optimal activity. However, the studies on the in vitro kinetic characterization of SOD from the rat liver cytosolic fraction have not been reported. The main objective of this study concerns the determination of the effect of three heavy metals such as arsenic, mercury, and lead on the activity of cytosolic SOD isolated from post nuclear supernatant (PNS) of rat liver. The activity of SOD was calculated using pyrogallol as a substrate. The stability and the sensitivity of enzyme activity were measured by assaying the enzyme activity at different temperature conditions. In order to determine the IC50 of the heavy metals, the enzyme activity was monitored in the presence of different concentrations of heavy metals. The values of all kinetic parameters including Km, Vmax, and Kcat were calculated by assaying SOD in the presence and absence of heavy metals. The results indicated that these heavy metals were able to significantly modulate the kinetic behavior of hepatic SOD. The data from present study could be utilized to develop suitable antidotes to mitigate the adverse effects of these heavy metals. Graphical Abstract.


Asunto(s)
Arsénico/metabolismo , Plomo/metabolismo , Mercurio/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Hígado/metabolismo , Masculino , Ratas
7.
J Endocrinol ; 244(1): 53-70, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31648182

RESUMEN

Estrogen deficiency reduces estrogen receptor-alpha (ERα) and promotes apoptosis in the hippocampus, inducing learning-memory deficits; however, underlying mechanisms remain less understood. Here, we explored the molecular mechanism in an ovariectomized (OVX) rat model, hypothesizing participation of autophagy and growth factor signaling that relate with apoptosis. We observed enhanced hippocampal autophagy in OVX rats, characterized by increased levels of autophagy proteins, presence of autophagosomes and inhibition of AKT-mTOR signaling. Investigating upstream effectors of reduced AKT-mTOR signaling revealed a decrease in hippocampal heparin-binding epidermal growth factor (HB-EGF) and p-EGFR. Moreover, 17ß-estradiol and HB-EGF treatments restored hippocampal EGFR activation and alleviated downstream autophagy process and neuronal loss in OVX rats. In vitro studies using estrogen receptor (ERα)-silenced primary hippocampal neurons further corroborated the in vivo observations. Additionally, in vivo and in vitro studies suggested the participation of an attenuated hippocampal neuronal HB-EGF and enhanced autophagy in apoptosis of hippocampal neurons in estrogen- and ERα-deficient conditions. Subsequently, we found evidence of mitochondrial loss and mitophagy in hippocampal neurons of OVX rats and ERα-silenced cells. The ERα-silenced cells also showed a reduction in ATP production and an HB-EGF-mediated restoration. Finally in concordance with molecular studies, inhibition of autophagy and treatment with HB-EGF in OVX rats restored cognitive performances, assessed through Y-Maze and passive avoidance tasks. Overall, our study, for the first time, links neuronal HB-EGF/EGFR signaling and autophagy with ERα and memory performance, disrupted in estrogen-deficient condition.


Asunto(s)
Receptor alfa de Estrógeno/deficiencia , Estrógenos/deficiencia , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Trastornos de la Memoria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Estradiol/farmacología , Femenino , Hipocampo/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Ovariectomía , Ratas , Transducción de Señal
8.
Toxicol Sci ; 159(1): 137-158, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28903487

RESUMEN

Arsenic promotes hippocampal neuronal damage inducing cognitive impairments. However, mechanism arbitrating arsenic-mediated cognitive deficits remains less-known. Here, we identified that chronic exposure to environmentally relevant doses of arsenic increased apoptosis, characterized by caspase-3 activation, poly(ADP-ribose) polymerase cleavage and Terminal deoxynucleotidyl transferase dUTP nick-end labeling of rat hippocampal neurons, marked by NeuN. Investigating apoptotic mechanism through invivo and invitro studies revealed that arsenic promoted bone morphogenetic protein-2 (BMP2) expression, supported by increased BMP-receptor2 (BMPR2) and p-Smad1/5 in hippocampal neurons. BMP2-silencing and treatment with BMP antagonist, noggin, attenuated the arsenic-induced apoptosis and loss in hippocampal neurons. We then investigated whether BMP2/Smad signaling stimulated neuronal apoptosis independently or required other intermediate pathways. We hypothesized participation of brain-derived neurotrophic factor (BDNF) that promotes neuronal survival. We identified an arsenic-mediated attenuation of BDNF-dependent TrkB signaling, and observed that co-treatment with recombinant-BDNF reinstated BDNF/TrkB and reduced neuronal apoptosis. To probe whether BMP2/Smad and BDNF/TrkB pathways could be linked, we co-treated arsenic with noggin or recombinant BDNF. We detected a noggin-mediated restored BDNF/TrkB, while recombinant-BDNF failed to affect BMP2/Smad signaling. In addition, we found that TrkB-inhibitor, K252a, nullified noggin-induced protection, proving the necessity of a downstream reduced BDNF/TrKB signaling for BMP2/Smad-mediated apoptosis in arsenic-treated neurons. We further related our observations with cognitive performances, and detected noggin-mediated restoration of transfer latency time and learning-memory ability for passive avoidance and Y-Maze tests respectively in arsenic-treated rats. Overall, our study proves that arsenic promotes hippocampal neuronal apoptosis through an up-regulated BMP2/Smad-dependent attenuation of BDNF/TrkB pathway, inducing cognitive deficits.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsénico/toxicidad , Proteína Morfogenética Ósea 2/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Trastornos del Conocimiento/inducido químicamente , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Receptor trkB/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Animales , Células Cultivadas , Femenino , Hipocampo/patología , Neuronas/patología , Embarazo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
9.
Neurobiol Dis ; 95: 179-93, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27431094

RESUMEN

Chronic cerebral hypoperfusion (CCH) manifests Alzheimer's Disease (AD) neuropathology, marked by increased amyloid beta (Aß). Besides, hypoxia stimulates Heparin-binding EGF-like growth factor (HB-EGF) mRNA expression in the hippocampus. However, involvement of HB-EGF in CCH-induced Aß pathology remains unidentified. Here, using Bilateral Common Carotid Artery Occlusion mouse model, we explored the mechanism of HB-EGF regulated Aß induction in CCH. We found that HB-EGF inhibition suppressed, while exogenous-HB-EGF triggered hippocampal Aß, proving HB-EGF-dependent Aß increase. We also detected that HB-EGF affected the expression of primary Aß transporters, receptor for advanced glycation end-products (RAGE) and lipoprotein receptor-related protein-1 (LRP-1), indicating impaired Aß clearance across the blood-brain barrier (BBB). An HB-EGF-dependent loss in BBB integrity supported impaired Aß clearance. The effect of HB-EGF on Amyloid Precursor Protein pathway was relatively insignificant, suggesting a lesser effect on Aß generation. Delving into BBB disruption mechanism demonstrated HB-EGF-mediated stimulation of Matrix metalloprotease-9 (MMP9), which affected BBB via HB-EGF-ectodomain shedding and epidermal growth factor receptor activation. Examining the intersection of HB-EGF-regulated pathway and hypoxia revealed HB-EGF-dependent increase in transcription factor, Hypoxia-inducible factor-1alpha (HIF1α). Further, via binding to hypoxia-responsive elements in MMP9 gene, HIF1α stimulated MMP9 expression, and therefore appeared as a prominent intermediary in HB-EGF-induced BBB damage. Overall, our study reveals the essential role of HB-EGF in triggering CCH-mediated Aß accumulation. The proposed mechanism involves an HB-EGF-dependent HIF1α increase, generating MMP9 that stimulates soluble-HB-EGF/EGFR-induced BBB disintegration. Consequently, CCH-mediated hippocampal RAGE and LRP-1 deregulation together with BBB damage impair Aß transport and clearance where HB-EGF plays a pivotal role.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Barrera Hematoencefálica/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Transporte Biológico/fisiología , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Masculino , Ratones , Perfusión , Receptor para Productos Finales de Glicación Avanzada/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...