Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 1, 2023 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-36593485

RESUMEN

Chronic hyperammonemia, a main contributor to hepatic encephalopathy (HE), leads to neuroinflammation which alters neurotransmission leading to cognitive impairment. There are no specific treatments for the neurological alterations in HE. Extracellular vesicles (EVs) from mesenchymal stem cells (MSCs) reduce neuroinflammation in some pathological conditions. The aims were to assess if treatment of hyperammonemic rats with EVs from MSCs restores cognitive function and analyze the underlying mechanisms. EVs injected in vivo reach the hippocampus and restore performance of hyperammonemic rats in object location, object recognition, short-term memory in the Y-maze and reference memory in the radial maze. Hyperammonemic rats show reduced TGFß levels and membrane expression of TGFß receptors in hippocampus. This leads to microglia activation and reduced Smad7-IkB pathway, which induces NF-κB nuclear translocation in neurons, increasing IL-1ß which alters AMPA and NMDA receptors membrane expression, leading to cognitive impairment. These effects are reversed by TGFß in the EVs from MSCs, which activates TGFß receptors, reducing microglia activation and NF-κB nuclear translocation in neurons by normalizing the Smad7-IkB pathway. This normalizes IL-1ß, AMPA and NMDA receptors membrane expression and, therefore, cognitive function. EVs from MSCs may be useful to improve cognitive function in patients with hyperammonemia and minimal HE.


Asunto(s)
Vesículas Extracelulares , Hiperamonemia , Células Madre Mesenquimatosas , Ratas , Animales , Ratas Wistar , Inflamación/metabolismo , Enfermedades Neuroinflamatorias , Receptores de N-Metil-D-Aspartato/metabolismo , Hiperamonemia/terapia , Hiperamonemia/metabolismo , FN-kappa B/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología , Hipocampo/metabolismo , Cognición , Células Madre Mesenquimatosas/metabolismo , Vesículas Extracelulares/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Int J Mol Sci ; 23(20)2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36293192

RESUMEN

Minimal hepatic encephalopathy (MHE) is associated with changes in the immune system including an increased pro-inflammatory environment and altered differentiation of CD4+ T lymphocytes. The mechanisms remain unknown. Changes in extracellular vesicle (EV) cargo including proteins and miRNAs could play a main role as mediators of immune system changes associated with MHE. The aim was to assess whether plasma EVs from MHE patients played a role in inducing the pro-inflammatory environment and altered differentiation of CD4+ T lymphocyte subtypes in MHE patients. We characterized the miRNA and protein cargo of plasma EVs from 50 cirrhotic patients (27 without and 23 with MHE) and 24 controls. CD4+ T cells from the controls were cultured with plasma EVs from the three groups of study, and the cytokine release and differentiation to CD4+ T-cell subtypes were assessed. Plasma EVs from MHE patients had altered miRNA and protein contents, and were enriched in inflammatory factors compared to the controls and patients without MHE. EVs from MHE patients modulated the expression of pro-inflammatory IL-17, IL-21, and TNF-α and anti-inflammatory TGF-ß in cultured CD4+ T lymphocytes, and increased the proportion of Th follicular and Treg cells and the activation of Th17 cells. In conclusion, plasma EVs could play an important role in the induction of immune changes observed in MHE.


Asunto(s)
Vesículas Extracelulares , Encefalopatía Hepática , MicroARNs , Humanos , Interleucina-17/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Vesículas Extracelulares/metabolismo , Citocinas/metabolismo , Linfocitos T Colaboradores-Inductores , MicroARNs/genética , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Cirrosis Hepática/metabolismo
3.
Neurochem Res ; 47(7): 2016-2031, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35386048

RESUMEN

Chronic hyperammonemia alters membrane expression of AMPA and NMDA receptors subunits in hippocampus leading to impaired memory and learning. Increasing extracellular cGMP normalizes these alterations. However, it has not been studied whether hyperammonemia alters the function of AMPA and NMDA receptors. The aims of this work were: (1) assess if hyperammonemia alters AMPA and NMDA receptors function; (2) analyze if extracellular cGMP reverses these alterations. A multielectrode array device was used to stimulate Schäffer collaterals and record postsynaptic currents in the CA1 region in hippocampal slices from control and hyperammonemic rats and analyze different features of the excitatory postsynaptic potentials. Hyperammonemia reduces the amplitude and delays appearance of AMPA EPSPs, whereas increases amplitude, hyperpolarization, depolarization and desensitization area of the NMDA EPSPs. These alterations in AMPA and NMDA function are accentuated as the stimulation intensity increases. Adding extracellular cGMP reverses the alteration in amplitude in both, AMPA and NMDA EPSPs. In control slices extracellular cGMP decreases the AMPA and NMDA EPSPs amplitude and delays the response of neurons and the return to the resting potential at all stimulation intensities. In conclusion, hyperammonemia decreases the AMPA response, whereas increases the NMDA response and extracellular cGMP reverses these alterations.


Asunto(s)
Hiperamonemia , Receptores de N-Metil-D-Aspartato , Animales , GMP Cíclico/metabolismo , Hipocampo/metabolismo , Hiperamonemia/metabolismo , N-Metilaspartato/metabolismo , N-Metilaspartato/farmacología , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico
4.
Vitam Horm ; 118: 247-288, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35180929

RESUMEN

Cyclic Guanosine-Monophosphate (cGMP) is implicated as second messenger in a plethora of pathways and its effects are executed mainly by cGMP-dependent protein kinases (PKG). It is involved in both peripheral (cardiovascular regulation, intestinal secretion, phototransduction, etc.) and brain (hippocampal synaptic plasticity, neuroinflammation, cognitive function, etc.) processes. Stimulation of hippocampal cGMP signaling have been proved to be beneficial in animal models of aging, Alzheimer's disease or hepatic encephalopathy, restoring different cognitive functions such as passive avoidance, object recognition or spatial memory. However, even when some inhibitors of cGMP-degrading enzymes (PDEs) are already used against peripheral pathologies, their utility as neurological treatments is still under clinical investigation. Additionally, it has been demonstrated a list of cGMP roles as not second but first messenger. The role of extracellular cGMP has been specially studied in hippocampal function and cognitive impairment in animal models and it has emerged as an important modulator of neuroinflammation-mediated cognitive alterations and hippocampal synaptic plasticity malfunction. Specifically, it has been demonstrated that extracellular cGMP decreases hippocampal IL-1ß levels restoring membrane expression of glutamate receptors in the hippocampus and cognitive function in hyperammonemic rats. The mechanisms implicated are still unclear and might involve complex interactions between hippocampal neurons, astrocytes and microglia. Membrane targets for extracellular cGMP are still poorly understood and must be addressed in future studies.


Asunto(s)
GMP Cíclico , Hiperamonemia , Animales , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Hipocampo/metabolismo , Hiperamonemia/metabolismo , Microglía/metabolismo , Ratas , Transducción de Señal/fisiología
5.
J Clin Med ; 10(2)2021 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-33440769

RESUMEN

Cirrhotic patients may experience alterations in the peripheral nervous system and in somatosensory perception. Impairment of the somatosensory system could contribute to cognitive and motor alterations characteristic of minimal hepatic encephalopathy (MHE), which affects up to 40% of cirrhotic patients. We assessed the relationship between MHE and alterations in thermal, vibration, and/or heat pain sensitivity in 58 cirrhotic patients (38 without and 20 with MHE according to Psychometric Hepatic Encephalopathy Score) and 39 controls. All participants underwent attention and coordination tests, a nerve conduction study, autonomic function testing, and evaluation of sensory thresholds (vibration, cooling, and heat pain detection) by electromyography and quantitative sensory testing. The detection thresholds for cold and heat pain on the foot were higher in patients with, than those without MHE. This hyposensitivity was correlated with attention deficits. Reaction times in the foot were longer in patients with, than without MHE. Patients with normal sural nerve amplitude showed altered thermal sensitivity and autonomic function, with stronger alterations in patients with, than in those without MHE. MHE patients show a general decrease in cognitive and sensory abilities. Small fibers of the autonomic nervous system and thermal sensitivity are altered early on in MHE, before large sensory fibers. Quantitative sensory testing could be used as a marker of MHE.

6.
J Neuroinflammation ; 17(1): 269, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917219

RESUMEN

BACKGROUND: Chronic hyperammonemia induces neuroinflammation in cerebellum, with glial activation and enhanced activation of the TNFR1-NF-kB-glutaminase-glutamate-GABA pathway. Hyperammonemia also increases glycinergic neurotransmission. These alterations contribute to cognitive and motor impairment. Activation of glycine receptors is reduced by extracellular cGMP, which levels are reduced in cerebellum of hyperammonemic rats in vivo. We hypothesized that enhanced glycinergic neurotransmission in hyperammonemic rats (1) contributes to induce neuroinflammation and glutamatergic and GABAergic neurotransmission alterations; (2) is a consequence of the reduced extracellular cGMP levels. The aims were to assess, in cerebellum of hyperammonemic rats, (a) whether blocking glycine receptors with the antagonist strychnine reduces neuroinflammation; (b) the cellular localization of glycine receptor; (c) the effects of blocking glycine receptors on the TNFR1-NF-kB-glutaminase-glutamate-GABA pathway and microglia activation; (d) whether adding extracellular cGMP reproduces the effects of strychnine. METHODS: We analyzed in freshly isolated cerebellar slices from control or hyperammonemic rats the effects of strychnine on activation of microglia and astrocytes, the content of TNFa and IL1b, the surface expression of ADAM17, TNFR1 and transporters, the phosphorylation levels of ERK, p38 and ADAM17. The cellular localization of glycine receptor was assessed by immunofluorescence. We analyzed the content of TNFa, IL1b, HMGB1, glutaminase, and the level of TNF-a mRNA and NF-κB in Purkinje neurons. Extracellular concentrations of glutamate and GABA were performed by in vivo microdialysis in cerebellum. We tested whether extracellular cGMP reproduces the effects of strychnine in ex vivo cerebellar slices. RESULTS: Glycine receptors are expressed mainly in Purkinje cells. In hyperammonemic rats, enhanced glycinergic neurotransmission leads to reduced membrane expression of ADAM17, resulting in increased surface expression and activation of TNFR1 and of the associated NF-kB pathway. This increases the expression in Purkinje neurons of TNFa, IL-1b, HMGB1, and glutaminase. Increased glutaminase activity leads to increased extracellular glutamate, which increases extracellular GABA. Increased extracellular glutamate and HMGB1 potentiate microglial activation. Blocking glycine receptors with strychnine or extracellular cGMP completely prevents the above pathway in hyperammonemic rats. CONCLUSIONS: Glycinergic neurotransmission modulates neuroinflammation. Enhanced glycinergic neurotransmission in hyperammonemia would be due to reduced extracellular cGMP. These results shed some light on possible new therapeutic target pathways for pathologies associated to neuroinflammation.


Asunto(s)
Cerebelo/efectos de los fármacos , Glicinérgicos/farmacología , Hiperamonemia/metabolismo , Receptores de Glicina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Estricnina/farmacología , Transmisión Sináptica/efectos de los fármacos , Proteína ADAM17/metabolismo , Animales , Cerebelo/metabolismo , GMP Cíclico/metabolismo , FN-kappa B/metabolismo , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Ratas , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transmisión Sináptica/fisiología
7.
Cells ; 9(3)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32121257

RESUMEN

Minimal hepatic encephalopathy is associated with changes in the peripheral immune system which are transferred to the brain, leading to neuroinflammation and thus to cognitive and motor impairment. Mechanisms by which changes in the immune system induce cerebral alterations remain unclear. Extracellular vesicles (EVs) seem to play a role in this process in certain pathologies. The aim of this work was to assess whether EVs play a role in the induction of neuroinflammation in cerebellum and motor incoordination by chronic hyperammonemia. We characterized the differences in protein cargo of EVs from plasma of hyperammonemic and control rats by proteomics and Western blot. We assessed whether injection of EVs from hyperammonemic to normal rats induces changes in neuroinflammation in cerebellum and motor incoordination similar to those exhibited by hyperammonemic rats. We found that hyperammonemia increases EVs amount and alters their protein cargo. Differentially expressed proteins are mainly associated with immune system processes. Injected EVs enter Purkinje neurons and microglia. Injection of EVs from hyperammonemic, but not from control rats, induces motor incoordination, which is mediated by neuroinflammation, microglia and astrocytes activation and increased IL-1b, TNFα, its receptor TNFR1, NF-kB in microglia, glutaminase I, and GAT3 in cerebellum. Plasma EVs from hyperammonemic rats carry molecules necessary and sufficient to trigger neuroinflammation in cerebellum and the mechanisms leading to motor incoordination.


Asunto(s)
Vesículas Extracelulares/metabolismo , Encefalopatía Hepática/inducido químicamente , Hiperamonemia/complicaciones , Trastornos de la Destreza Motora/inducido químicamente , Enfermedades del Sistema Nervioso/inducido químicamente , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Masculino , Ratas , Ratas Wistar
8.
J Hepatol ; 73(3): 582-592, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-30654069

RESUMEN

BACKGROUND & AIMS: Chronic hyperammonemia induces neuroinflammation which mediates cognitive impairment. How hyperammonemia induces neuroinflammation remains unclear. We aimed to assess whether: chronic hyperammonemia induces peripheral inflammation, and whether this then contributes to neuroinflammation, altered neurotransmission and impaired spatial learning - before assessing whether this neuroinflammation and impairment is reversible following hyperammonemia elimination or treatment of peripheral inflammation with anti-TNF-α. METHODS: Chronic hyperammonemia was induced by feeding rats an ammonia-containing diet. Peripheral inflammation was analyzed by measuring PGE2, TNF-α, IL-6 and IL-10. We tested whether chronic anti-TNF-α treatment improves peripheral inflammation, neuroinflammation, membrane expression of glutamate receptors in the hippocampus and spatial learning. RESULTS: Hyperammonemic rats show a rapid and reversible induction of peripheral inflammation, with increased pro-inflammatory PGE2, TNF-α and IL-6, followed at around 10 days by reduced anti-inflammatory IL-10. Peripheral anti-TNF-α treatment prevents peripheral inflammation induction and the increase in IL-1b and TNF-α and microglia activation in hippocampus of the rats, which remain hyperammonemic. This is associated with prevention of the altered membrane expression of glutamate receptors and of the impairment of spatial memory assessed in the radial and Morris water mazes. CONCLUSIONS: This report unveils a new mechanism by which chronic hyperammonemia induces neurological alterations: induction of peripheral inflammation. This suggests that reducing peripheral inflammation by safe procedures would improve cognitive function in patients with minimal hepatic encephalopathy. LAY SUMMARY: This article unveils a new mechanism by which chronic hyperammonemia induces cognitive impairment in rats: chronic hyperammonemia per se induces peripheral inflammation, which mediates many of its effects on the brain, including induction of neuroinflammation, which alters neurotransmission, leading to cognitive impairment. It is also shown that reducing peripheral inflammation by treating rats with anti-TNF-α, which does not cross the blood-brain barrier, prevents hyperammonemia-induced neuroinflammation, alterations in neurotransmission and cognitive impairment.


Asunto(s)
Antiinflamatorios/administración & dosificación , Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Hiperamonemia/complicaciones , Infliximab/administración & dosificación , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Disfunción Cognitiva/sangre , Modelos Animales de Enfermedad , Encefalopatía Hepática/tratamiento farmacológico , Encefalopatía Hepática/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratas , Ratas Wistar , Aprendizaje Espacial/efectos de los fármacos , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/sangre
9.
Sci Rep ; 9(1): 17119, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31745217

RESUMEN

Patients with Benign prostatic hyperplasia, low urinary tract symptoms, and erectile dysfunction (BPH/LUTS-ED) present chronic inflammation. We studied in patients with BPH/LUTS-ED the effect of tadalafil treatment (5 mg/day) on changes in peripheral inflammation, cognitive function, and the auditory evoked potential, "mismatch negativity" (MMN). Nine patients with BPH/LUTS-ED and 12 controls performed psychometric tests, MMN. IL-6, IL-17, IL-18, cGMP and CD4+CD28- autoreactive T-cells were measured in blood. Patients with BPH/LUTS-ED performed psychometric tests, MMN, and blood extraction at baseline and after tadalafil treatment. Patients with BPH/LUTS-ED showed increased CD4+CD28- autoreactive T-cells (p < 0.05), and higher levels of pro-inflammatory interleukins IL-6 (p < 0.001), IL-17 and IL-18 (p < 0.05), compared to controls. Patients got lower scores than controls in psychometric tests assessing mental processing speed and attention (p < 0.05), and showed lower amplitude (p < 0.01) and area (p < 0.05) of MMN wave than controls. Inflammatory, psychometric and electrophysiological parameters were normalized after tadalafil treatment. In conclusion, there is a pro-inflammatory environment in blood in patients with BPH/LUTS-ED which would induce cognitive impairment and alter MMN. Phosphodiesterase-5 inhibition with tadalafil exerts anti-inflammatory effects and ameliorates cognitive function and MMN parameters. Tadalafil could be a promising candidate for chronic treatment in other inflammatory pathologies associated with mild cognitive impairment.


Asunto(s)
Cognición/efectos de los fármacos , Disfunción Eréctil/tratamiento farmacológico , Inflamación/prevención & control , Síntomas del Sistema Urinario Inferior/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Hiperplasia Prostática/tratamiento farmacológico , Tadalafilo/uso terapéutico , Anciano , Disfunción Eréctil/patología , Humanos , Síntomas del Sistema Urinario Inferior/patología , Masculino , Persona de Mediana Edad , Hiperplasia Prostática/patología , Resultado del Tratamiento
10.
J Transl Med ; 17(1): 293, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31462286

RESUMEN

BACKGROUND: Minimal hepatic encephalopathy (MHE) in cirrhotic patients is associated with specific changes in parameters of the immune system reflecting a more pro-inflammatory environment than in patients without MHE. The aims of this work were to assess the effects of rifaximin treatment of cirrhotic patients with MHE on: (1) MHE; (2) intermediate (CD14++CD16+) pro-inflammatory monocytes; (3) expression of early activation marker CD69 in T lymphocytes; (4) autoreactive CD4+CD28- T lymphocytes; (5) differentiation of CD4+ T lymphocytes to Th follicular and Th22; (6) serum IgG levels; and (7) levels of some pro-inflammatory cytokines. METHODS: These parameters were measured by immunophenotyping and cytokine profile analysis in 30 controls without liver disease, 30 cirrhotic patients without MHE and 22 patients with MHE. Patients with MHE were treated with rifaximin and the same parameters were measured at 3 and 6 months of treatment. We assessed if changes in these parameters are different in patients who improve MHE (responders) and those who remain in MHE (non-responders). RESULTS: Rifaximin improved MHE in 59% of patients with MHE. In these responder patients rifaximin normalized all alterations in the immune system measured while in non-responders it normalizes only IL-6, CCL20, and differentiation of T lymphocytes to Th22. Non-responder patients do not show increased expression of CD69 before treatment. CONCLUSIONS: Rifaximin normalizes changes in the immune system in patients who improve MHE but not in non-responders. Some alterations before treatment are different in responders and non-responders. Understanding these differences may identify predictors of the response of MHE to rifaximin.


Asunto(s)
Encefalopatía Hepática/tratamiento farmacológico , Encefalopatía Hepática/inmunología , Inmunofenotipificación , Rifaximina/uso terapéutico , Citocinas/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Encefalopatía Hepática/sangre , Humanos , Inmunoglobulina G/sangre , Monocitos/efectos de los fármacos , Psicometría , Rifaximina/farmacología , Linfocitos T/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
FASEB J ; 33(9): 9913-9928, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31162953

RESUMEN

Activated microglia and increased brain IL-1ß play a main role in cognitive impairment in much pathology. We studied the role of IL-1ß in neuroinflammation-induced impairment of the following different types of learning and memory: novel object recognition (NOR), novel object location (NOL), spatial learning, reference memory (RM), and working memory (WM). All these processes are impaired in hyperammonemic rats. We assessed which of these types of learning and memory are restored by blocking the IL-1 receptor in vivo in hyperammonemic rats and the possible mechanisms involved. Blocking the IL-1 receptor reversed microglial activation in the hippocampus, perirhinal cortex, and prefrontal cortex but not in the postrhinal cortex. This was associated with the restoration of NOR and WM but not of tasks involving a spatial component (NOL and RM). This suggests that IL-1ß would be involved in neuroinflammation-induced nonspatial memory impairment, whereas spatial memory impairment would be IL-1ß-independent and would be mediated by other proinflammatory factors.-Taoro-González, L., Cabrera-Pastor, A., Sancho-Alonso, M., Arenas, Y. M., Meseguer-Estornell, F., Balzano, T., ElMlili, N., Felipo, V. Differential role of interleukin-1ß in neuroinflammation-induced impairment of spatial and nonspatial memory in hyperammonemic rats.


Asunto(s)
Hiperamonemia/inducido químicamente , Inflamación/inducido químicamente , Interleucina-1beta/metabolismo , Memoria/efectos de los fármacos , Amoníaco/administración & dosificación , Amoníaco/toxicidad , Alimentación Animal , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Microglía/efectos de los fármacos , Microglía/fisiología , Subunidades de Proteína , Ratas , Ratas Wistar , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de GABA/genética , Receptores de GABA/metabolismo , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
12.
Acta Physiol (Oxf) ; 226(2): e13270, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30830722

RESUMEN

Several million patients with liver cirrhosis suffer minimal hepatic encephalopathy (MHE), with mild cognitive and coordination impairments that reduce their quality of life and life span. Hyperammonaemia and peripheral inflammation act synergistically to induce these neurological alterations. We propose that MHE appearance is because of the changes in peripheral immune system, which are transmitted to brain, leading to neuroinflammation that alters neurotransmission leading to cognitive and motor alterations. We summarize studies showing that MHE in cirrhotic patients is associated with alterations in the immune system and that patients died with HE show neuroinflammation in cerebellum, with microglial and astrocytic activation and Purkinje cell loss. We also summarize studies in animal models of MHE on the role of peripheral inflammation in neuroinflammation induction, how neuroinflammation alters neurotransmission and how this leads to cognitive and motor alterations. These studies identify therapeutic targets and treatments that improve cognitive and motor function. Rats with MHE show neuroinflammation in hippocampus and altered NMDA and AMPA receptor membrane expression, which impairs spatial learning and memory. Neuroinflammation in cerebellum is associated with altered GABA transporters and extracellular GABA, which impair motor coordination and learning in a Y maze. These alterations are reversed by treatments that reduce peripheral inflammation (anti-TNFα, ibuprofen), neuroinflammation (sulphoraphane, p38 inhibitors), GABAergic tone (bicuculline, pregnenolone sulphate) or increase extracellular cGMP (sildenafil or cGMP). The mechanisms identified would also occur in other chronic diseases associated with inflammation, aging and some mental and neurodegenerative diseases. Treatments that improve MHE may also be beneficial to treat these pathologies.


Asunto(s)
Cognición/fisiología , Encefalopatía Hepática/metabolismo , Inflamación/metabolismo , Actividad Motora/fisiología , Transmisión Sináptica/fisiología , Animales , Encefalopatía Hepática/fisiopatología , Humanos , Hiperamonemia/metabolismo
13.
Neuropharmacology ; 161: 107496, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30641078

RESUMEN

Trafficking of glutamate, glutamine and GABA between astrocytes and neurons is essential to maintain proper neurotransmission. Chronic hyperammonemia alters neurotransmission and cognitive function. The aims of this work were to analyze in cerebellum of rats the effects of chronic hyperammonemia on: a) extracellular glutamate, glutamine and GABA concentrations; b) membrane expression of glutamate, glutamine and GABA transporters; c) how they are modulated by extracellular cGMP. Hyperammonemic rats show increased levels of extracellular glutamate, glutamine, GABA and citrulline in cerebellum in vivo. Hyperammonemic rats show: a) increased membrane expression of the astrocytic glutamine transporter SNAT3 and reduced membrane expression of the neuronal transporter SNAT1; b) reduced membrane expression of the neuronal GABA transporter GAT1 and increased membrane expression of the astrocytic GAT3 transporter; c) reduced membrane expression of the astrocytic glutamate transporters GLAST and GLT-1 and of the neuronal transporter EAAC1. Increasing extracellular cGMP normalizes membrane expression of SNAT3, GAT3, GAT1 and GLAST and extracellular glutamate, glutamine, GABA and citrulline hyperammonemic rats. Extracellular cGMP also modulates membrane expression of most transporters in control rats, reducing membrane expression of SNAT1, GLT-1 and EAAC1 and increasing that of GAT1 and GAT3. Modulation of SNAT3, SNAT1, GLT-1 and EAAC1 by extracellular cGMP would be mediated by inhibition of glycine receptors. These data suggest that, in pathological situations such as hyperammonemia, hepatic encephalopathy or Alzheimer's disease, reduced levels of extracellular cGMP contribute to alterations in membrane expression of glutamine, glutamate and GABA transporters, in the extracellular levels of glutamine, glutamate and GABA and in neurotransmission. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Asunto(s)
Membrana Celular/metabolismo , Cerebelo/metabolismo , GMP Cíclico/farmacología , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Hiperamonemia/metabolismo , Proteínas de Transporte de Neurotransmisores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Citrulina/metabolismo , Espacio Extracelular , Masculino , Proteínas de Transporte de Neurotransmisores/fisiología , Ratas , Ratas Wistar , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
14.
Mol Neurobiol ; 56(6): 4428-4439, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30328550

RESUMEN

Chronic hyperammonemia impairs spatial memory by altering membrane expression of GluA1 and GluA2 subunits of AMPA receptors in hippocampus. Intracerebral administration of extracellular cGMP to hyperammonemic rats restores spatial memory and membrane expression of AMPA receptors. The underlying molecular mechanisms remain unknown and cannot be analyzed in vivo. The aims of the present work were to (1) assess whether extracellular cGMP reverses the alterations in membrane expression of GluA1 and GluA2 in hippocampus of hyperammonemic rats ex vivo and (2) identify the underlying mechanisms. To reach these aims, we used freshly isolated hippocampal slices from control and hyperammonemic rats and treated them ex vivo with extracellular cGMP. Extracellular cGMP normalizes membrane expression of GluA2 restoring its phosphorylation in Ser880 because it restores PKCζ activation by Thr560 auto-phosphorylation, which is a consequence of normalization by extracellular cGMP of phosphorylation and activity of p38 which was increased in hyperammonemic rats. Normalization of p38 is a consequence of normalization of membrane expression of the GluN2B subunit of NMDA receptor, mediated by a reduction in its phosphorylation in Tyr1472 due to reduction of Src activation, which was over-activated in hyperammonemic rats. Extracellular cGMP also restores membrane expression of GluA1 increasing its phosphorylation at Ser831 because it restores CaMKII membrane association and phosphorylation in Thr286. All these effects of extracellular cGMP are due to a reduction of hippocampal IL-1ß levels in hyperammonemic rats, which reduces IL-1 receptor-mediated Src over-activation. Reduction in IL-1ß levels is due to the reduction of microglia activation in hippocampus of hyperammonemic rats.


Asunto(s)
Membrana Celular/metabolismo , GMP Cíclico/farmacología , Espacio Extracelular/química , Hipocampo/metabolismo , Hiperamonemia/metabolismo , Receptores AMPA/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Membrana Celular/efectos de los fármacos , Interleucina-1beta/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Biológicos , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Subunidades de Proteína/metabolismo , Ratas Wistar , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/metabolismo
15.
J Neuroinflammation ; 15(1): 36, 2018 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-29422059

RESUMEN

BACKGROUND: Hyperammonemic rats reproduce the cognitive alterations of patients with hepatic encephalopathy, including altered spatial memory, attributed to altered membrane expression of AMPA receptor subunits in hippocampus. Neuroinflammation mediates these cognitive alterations. We hypothesized that hyperammonemia-induced increase in IL-1ß in hippocampus would be responsible for the altered GluA1 and GluA2 membrane expression. The aims of this work were to (1) assess if increased IL-1ß levels and activation of its receptor are responsible for the changes in GluA1 and/or GluA2 membrane expression in hyperammonemia and (2) identify the mechanisms by which activation of IL-1 receptor leads to altered membrane expression of GluA1 and GluA2. METHODS: We analyzed in hippocampal slices from control and hyperammonemic rat membrane expression of AMPA receptors using the BS3 cross-linker and phosphorylation of the GluA1 and GluA2 subunits using phosphor-specific antibodies. The IL-1 receptor was blocked with IL-Ra, and the signal transduction pathways involved in modulation of membrane expression of GluA1 and GluA2 were analyzed using inhibitors of key steps. RESULTS: Hyperammonemia reduces GluA1 and increases GluA2 membrane expression and reduces phosphorylation of GluA1 at Ser831 and of GluA2 at Ser880. Hyperammonemia increases IL-1ß, enhancing activation of IL-1 receptor. This leads to activation of Src. The changes in membrane expression of GluA1 and GluA2 are reversed by blocking the IL-1 receptor with IL-1Ra or by inhibiting Src with PP2. After Src activation, the pathways for GluA2 and GluA1 diverge. Src increases phosphorylation of GluN2B at Tyr14721 and membrane expression of GluN2B in hyperammonemic rats, leading to activation of MAP kinase p38, which binds to and reduces phosphorylation at Thr560 and activity of PKCζ, resulting in reduced phosphorylation at Ser880 and enhanced membrane expression of GluA2. Increased Src activity in hyperammonemic rats also activates PKCδ which enhances phosphorylation of GluN2B at Ser1303, reducing membrane expression of CaMKII and phosphorylation at Ser831 and membrane expression of GluA1. CONCLUSIONS: This work identifies two pathways by which neuroinflammation alters glutamatergic neurotransmission in hippocampus. The steps of the pathways identified could be targets to normalize neurotransmission in hyperammonemia and other pathologies associated with increased IL-1ß by acting, for example, on p38 or PKCδ. IL-1ß alters membrane expression of GluA1 and GluA2 AMPA receptor subunits by two difrerent mechanisms in the hippocampus of hyperammonemic rats.


Asunto(s)
Membrana Celular/metabolismo , Hipocampo/metabolismo , Hiperamonemia/metabolismo , Receptores AMPA/biosíntesis , Receptores Tipo I de Interleucina-1/metabolismo , Acetatos/toxicidad , Animales , Membrana Celular/efectos de los fármacos , Expresión Génica , Hipocampo/efectos de los fármacos , Hiperamonemia/inducido químicamente , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Ratas , Ratas Wistar , Receptores AMPA/genética , Receptores Tipo I de Interleucina-1/antagonistas & inhibidores
16.
Brain Behav Immun ; 69: 386-398, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29288802

RESUMEN

Hyperammonemia is a main contributor to cognitive impairment and motor in-coordination in patients with hepatic encephalopathy. Hyperammonemia-induced neuroinflammation mediates the neurological alterations in hepatic encephalopathy. Intracerebral administration of extracellular cGMP restores some but not all types of cognitive impairment. Motor in-coordination, is mainly due to increased GABAergic tone in cerebellum. We hypothesized that extracellular cGMP would restore motor coordination in hyperammonemic rats by normalizing GABAergic tone in cerebellum and that this would be mediated by reduction of neuroinflammation. The aims of this work were to assess whether chronic intracerebral administration of cGMP to hyperammonemic rats: 1) restores motor coordination; 2) reduces neuroinflammation in cerebellum; 3) reduces extracellular GABA levels and GABAergic tone in cerebellum; and also 4) to provide some advance in the understanding on the molecular mechanisms involved. The results reported show that rats with chronic hyperammonemia show neuroinflammation in cerebellum, including microglia and astrocytes activation and increased levels of IL-1b and TNFa and increased membrane expression of the TNFa receptor. This is associated with increased glutaminase expression and extracellular glutamate, increased amount of the GABA transporter GAT-3 in activated astrocytes, increased extracellular GABA in cerebellum and motor in-coordination. Chronic intracerebral administration of extracellular cGMP to rats with chronic hyperammonemia reduces neuroinflammation, including microglia and astrocytes activation and membrane expression of the TNFa receptor. This is associated with reduced nuclear NF-κB, glutaminase expression and extracellular glutamate, reduced amount of the GABA transporter GAT-3 in activated astrocytes and reduced extracellular GABA in cerebellum and restoration of motor coordination. The data support that extracellular cGMP restores motor coordination in hyperammonemic rats by reducing microglia activation and neuroinflammation, leading to normalization of extracellular glutamate and GABA levels in cerebellum and of motor coordination.


Asunto(s)
Cerebelo/metabolismo , GMP Cíclico/farmacología , Hiperamonemia/metabolismo , Inflamación/metabolismo , Destreza Motora/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Astrocitos/metabolismo , Bicuculina/farmacología , Cerebelo/efectos de los fármacos , Antagonistas de Receptores de GABA-A/farmacología , Glutaminasa/metabolismo , Masculino , Microglía/metabolismo , Destreza Motora/efectos de los fármacos , Ratas , Ratas Wistar
17.
ACS Chem Neurosci ; 9(2): 369-380, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29094921

RESUMEN

Exposure to pesticides has been associated with neurodevelopmental toxicity. Usually people are exposed to mixtures of pesticides. However, most studies analyze the effects of individual pesticides. Developmental exposure to mixtures of pesticides may result in additive effects or in antagonistic or synergistic effects. The aim of this work was to compare the effects of developmental exposure of rats to cypermethrin or endosulfan with the effects of its mixture on cognitive and motor function and on some underlying mechanisms. Exposure to individual pesticides or the mixture was from gestational day 7 to postnatal day 21. We analyzed the effects, in males and females, on spatial learning and memory, associative learning, anxiety, motor coordination, and spontaneous motor activity. We also analyzed neuroinflammation and NMDA receptor subunits in hippocampus and extracellular GABA in cerebellum. Exposure to the mixture, but not to individual pesticides, impaired spatial memory in males, associative learning in females, and increased motor activity in males and females. This indicates a synergistic effect of cypermethrin and endolsufan exposure on these end points. In contrast, motor coordination was impaired by individual exposure to endosulfan or cypermethrin, associated with increased extracellular GABA in cerebellum, but these effects were prevented in rats exposed to the mixture, indicating an antagonistic effect of cypermethrin and endolsufan exposure on these end points. The results show different interaction modes (synergism or antagonism) of the pesticides, depending on the end point analyzed and the sex of the rats.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Endosulfano/toxicidad , Plaguicidas/toxicidad , Piretrinas/toxicidad , Animales , Ansiedad/inducido químicamente , Ansiedad/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo , Antagonismo de Drogas , Sinergismo Farmacológico , Endosulfano/antagonistas & inhibidores , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Femenino , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Plaguicidas/antagonistas & inhibidores , Piretrinas/antagonistas & inhibidores , Ratas Wistar , Caracteres Sexuales , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Ácido gamma-Aminobutírico/metabolismo
18.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 286-295, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29107806

RESUMEN

Hyperammonemia contributes to altered neurotransmission and cognition in patients with hepatic encephalopathy. Hyperammonemia in rats affects differently high- and low-affinity AMPA receptors (AMPARs) in cerebellum. We hypothesized that hyperammonemia would alter differently membrane expression of AMPARs GluA1 and GluA2 subunits by altering its phosphorylation. This work aims were: 1) assess if hyperammonemia alters GluA1 and GluA2 subunits membrane expression in cerebellum and 2) analyze the underlying mechanisms. Hyperammonemia reduces membrane expression of GluA2 and enhances membrane expression of GluA1 in vivo. We show that changes in GluA2 and GluA1 membrane expression in hyperammonemia would be due to enhanced NMDA receptors activation which reduces cGMP levels and phosphodiesterase 2 (PDE2) activity, resulting in increased cAMP levels. This leads to increased protein kinase A (PKA) activity which activates phospholipase C (PLC) and protein kinase C (PKC) thus increasing phosphorylation of GluA2 in Ser880, which reduces GluA2 membrane expression, and phosphorylation of GluA1 in Ser831, which increases GluA1 membrane expression. Blocking NMDA receptors or inhibiting PKA, PLC or PKC normalizes GluA2 and GluA1 phosphorylation and membrane expression in hyperammonemic rats. Altered GluA2 and GluA1 membrane expression would alter signal transduction which may contribute to cognitive and motor alterations in hyperammonemia and hepatic encephalopathy.


Asunto(s)
Membrana Celular/metabolismo , Hiperamonemia/genética , Receptores AMPA/genética , Animales , Membrana Celular/patología , Enfermedad Crónica , Encefalopatía Hepática/genética , Encefalopatía Hepática/metabolismo , Encefalopatía Hepática/patología , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Transducción de Señal/genética , Transmisión Sináptica/genética
19.
Sci Rep ; 7(1): 17656, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29247190

RESUMEN

There is increasing evidence that extracellular cGMP modulates glutamatergic neurotransmission and some forms of learning. However, the underlying mechanisms remain unknown. We proposed the hypotheses that extracellular cGMP may regulate membrane expression of AMPA receptors. To do this extracellular cGMP should act on a membrane protein and activate signal transduction pathways modulating phosphorylation of the GluA1 and/or GluA2 subunits. It has been shown that extracellular cGMP modulates glycine receptors. The aims of this work were to assess: 1) whether extracellular cGMP modulates membrane expression of GluA1 and GluA2 subunits of AMPA receptors in cerebellum in vivo; 2) whether this is mediated by glycine receptors; 3) the role of GluA1 and GluA2 phosphorylation and 4) identify steps of the intracellular pathways involved. We show that extracellular cGMP modulates membrane expression of GluA1 and GluA2 in cerebellum in vivo and unveil the mechanisms involved. Extracellular cGMP reduced glycine receptor activation, modulating cAMP, protein kinases and phosphatases, and GluA1 and GluA2 phosphorylation, resulting in increased GluA1 and reduced GluA2 membrane expression. Extracellular cGMP therefore modulates membrane expression of AMPA receptors and glutamatergic neurotransmission. The steps identified may be therapeutic targets to improve neurotransmission and neurological function in pathological situations with abnormal glutamatergic neurotransmission.


Asunto(s)
Cerebelo/metabolismo , Proteínas de la Membrana/metabolismo , Subunidades de Proteína/metabolismo , Células de Purkinje/fisiología , Receptores AMPA/metabolismo , Animales , Cerebelo/patología , GMP Cíclico/metabolismo , Espacio Extracelular/metabolismo , Regulación de la Expresión Génica , Masculino , Proteínas de la Membrana/genética , Fosforilación , Subunidades de Proteína/genética , Ratas , Ratas Wistar , Receptores AMPA/genética , Receptores de Glicina/metabolismo , Transducción de Señal , Transmisión Sináptica
20.
CNS Neurosci Ther ; 23(5): 386-394, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28296282

RESUMEN

AIMS: Patients with liver disease may develop hepatic encephalopathy (HE), with cognitive impairment and motor in-coordination. Rats with HE due to portacaval shunts (PCS) show motor in-coordination. We hypothesized that in PCS rats: (i) Motor in-coordination would be due to enhanced GABAergic tone in cerebellum; (ii) increased GABAergic tone would be due to neuroinflammation; (iii) increasing cGMP would reduce neuroinflammation and GABAergic tone and restore motor coordination. To assess these hypotheses, we assessed if (i) treatment with sildenafil reduces neuroinflammation; (ii) reduced neuroinflammation is associated with reduced GABAergic tone and restored motor coordination. METHODS: Rats were treated with sildenafil to increase cGMP. Microglia and astrocytes activation were analyzed by immunohistochemistry, extracellular GABA by microdialysis, and motor coordination in the beam walking. RESULTS: PCS rats show neuroinflammation in cerebellum, with microglia and astrocytes activation, increased IL-1b and TNF-a and reduced YM-1 and IL-4. Membrane expression of the GABA transporter GAT1 is reduced, while GAT3 is increased. Extracellular GABA and motor in-coordination are increased. Sildenafil treatment eliminates neuroinflammation, microglia and astrocytes activation; changes in membrane expression of GABA transporters; and restores motor coordination. CONCLUSIONS: This study supports an interplay between cGMP-neuroinflammation and GABAergic neurotransmission in impairing motor coordination in PCS rats.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Cerebelo/efectos de los fármacos , Encefalopatía Hepática/tratamiento farmacológico , Destreza Motora/efectos de los fármacos , Citrato de Sildenafil/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Astrocitos/fisiología , Cerebelo/inmunología , Cerebelo/patología , Modelos Animales de Enfermedad , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Encefalopatía Hepática/patología , Encefalopatía Hepática/fisiopatología , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/patología , Microglía/fisiología , Destreza Motora/fisiología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA