Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J Neurochem ; 168(9): 2561-2576, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38676340

RESUMEN

Rapid eye movement sleep (REMS) maintains brain excitability at least by regulating Na-K ATPase activity. Although REMS deprivation (REMSD)-associated elevated noradrenaline (NA) increases Na-K ATPase protein expression, its mRNA transcription did not increase. We hypothesized and confirmed both in vivo as well as in vitro that elevated mRNA stability explains the apparent puzzle. The mRNA stability was measured in control and REMSD rat brain with or without in vivo treatment with α1-adrenoceptor (AR) antagonist, prazosin (PRZ). Upon REMSD, Na-K ATPase α1-, and α2-mRNA stability increased significantly, which was prevented by PRZ. To decipher the molecular mechanism of action, we estimated NA-induced Na-K ATPase mRNA stability in Neuro-2a cells under controlled conditions and by transcription blockage using Actinomycin D (Act-D). NA increased Na-K ATPase mRNA stability, which was prevented by PRZ and propranolol (PRP, ß-AR antagonist). The knockdown assay confirmed that the increased mRNA stabilization was induced by elevated cytoplasmic abundance of Human antigen R (HuR) and involving (Phospholipase C) PLC-mediated activation of Protein Kinase C (PKC). Additionally, using cell-impermeable Enz-link sulfo NHS-SS-Biotin, we observed that NA increased Na-K ATPase α1-subunits on the Neuro-2a cell surface. We conclude that REMSD-associated elevated NA, acting on α1- and ß-AR, increases nucleocytoplasmic translocation of HuR and increases Na-K ATPase mRNA stability, resulting in increased Na-K ATPase protein expression. The latter then gets translocated to the neuronal membrane surface involving both PKC and (Protein Kinase A) PKA-mediated pathways. These findings may be exploited for the amelioration of REMSD-associated chronic disorders and symptoms.


Asunto(s)
Proteína 1 Similar a ELAV , Norepinefrina , Proteína Quinasa C , Estabilidad del ARN , ATPasa Intercambiadora de Sodio-Potasio , Animales , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , Ratas , Norepinefrina/metabolismo , Proteína Quinasa C/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Proteína 1 Similar a ELAV/genética , Estabilidad del ARN/efectos de los fármacos , Masculino , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Mensajero/biosíntesis , Ratones , Transducción de Señal/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Línea Celular Tumoral , Ratas Wistar , Humanos
2.
Cell Mol Neurobiol ; 43(7): 3061-3080, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37165139

RESUMEN

Scaffold proteins Striatin and SG2NA assemble kinases and phosphatases into the signalling complexes called STRIPAK. Dysfunctional STRIPAKs cause cancer, cerebral cavernous malformations, etc. DJ-1, a sensor for oxidative stress, has long been associated with the Parkinson's disease, cancer, and immune disorders. SG2NA interacts with DJ-1 and Akt providing neuroprotection under oxidative stress. To dissect the role of SG2NA and DJ-1 in neuronal pathobiology, rat midbrain extracts were immunoprecipitated with SG2NA and sixty-three interacting proteins were identified. BN-PAGE followed by the LC-MS/MS showed 1030 comigrating proteins as the potential constituents of the multimeric complexes formed by SG2NA. Forty-three proteins were common between those identified by co-immunoprecipitation and the BN-PAGE. Co-immunoprecipitation with DJ-1 identified 179 interacting partners, of which forty-one also interact with SG2NA. Among those forty-one proteins immunoprecipitated with both SG2NA and DJ-1, thirty-nine comigrated with SG2NA in the BN-PAGE, and thus are bonafide constituents of the supramolecular assemblies comprising both DJ-1 and SG2NA. Among those thirty-nine proteins, seven are involved in mitochondrial oxidative phosphorylation. In rotenone-treated rats having Parkinson's like symptoms, the levels of both SG2NA and DJ-1 increased in the mitochondria; and the association of SG2NA with the electron transport complexes enhanced. In the hemi-Parkinson's model, where the rats were injected with 6-OHDA into the midbrain, the occupancy of SG2NA and DJ-1 in the mitochondrial complexes also increased. Our study thus reveals a new family of potential STRIPAK assemblies involving both SG2NA and DJ-1, with key roles in protecting midbrain from the oxidative stress.


Asunto(s)
Neoplasias , Enfermedad de Parkinson , Animales , Ratas , Cromatografía Liquida , Electrones , Mesencéfalo , Estrés Oxidativo , Espectrometría de Masas en Tándem
3.
BMC Neurol ; 23(1): 283, 2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507691

RESUMEN

The wakefulness-sleep cycle sets the pace of our life. Sleep research examines the transition between wakefulness and sleep, its hormonal regulation, and its pathological disruption. Understanding sleep mechanisms would improve quality-of-life well beyond sleep itself. To this aim, we invite contributions for the Collection "sleep physiology and circadian rhythms".


Asunto(s)
Ritmo Circadiano , Sueño , Humanos , Sueño/fisiología , Ritmo Circadiano/fisiología , Vigilia/fisiología , Calidad de Vida
4.
Eur J Neurosci ; 53(8): 2870-2900, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33529409

RESUMEN

Diabetes increases the likelihood of developing depression and vice versa. Research on this bidirectional association has somewhat managed to delineate the interplay among implicated physiological processes. Still, further exploration is required in this context. This review addresses the comorbidity by investigating suspected common pathophysiological mechanisms. One such factor is psychological stress which disturbs the hypothalamic-pituitary-adrenal axis causing hormonal imbalance. This includes elevated cortisol levels, a common biomarker of both depression and diabetes. Disrupted insulin signaling drives the hampered neurotransmission of serotonin, dopamine, and norepinephrine. Also, adipokine hormones such as adiponectin, leptin, and resistin and the orexigenic hormone, ghrelin, are involved in both depression and T2DM. This disarray further interferes with physiological processes encompassing sleep, the gut-brain axis, metabolism, and mood stability. Behavioral coping mechanisms, such as unhealthy eating, mediate disturbed glucose homeostasis, and neuroinflammation. This is intricately linked to oxidative stress, redox imbalance, and mitochondrial dysfunction. However, interventions such as psychotherapy, physical exercise, fecal microbiota transplantation, and insulin-sensitizing agents can help to manage the distressing condition. The possibility of glucagon-like peptide 1 possessing a therapeutic role has also been discussed. Nonetheless, there stands an urgent need for unraveling new correlating targets and biological markers for efficient treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbiota , Depresión/terapia , Diabetes Mellitus Tipo 2/terapia , Ejercicio Físico , Trasplante de Microbiota Fecal , Humanos , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Psicoterapia
5.
J Neurosci Res ; 99(7): 1815-1834, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33819353

RESUMEN

Rapid eye movement sleep (REMS) favors brain development and memory, while it is decreased in neurodegenerative diseases. REMS deprivation (REMSD) affects several physiological processes including memory consolidation; however, its detailed mechanism(s) of action was unknown. REMS reduces, while REMSD elevates noradrenaline (NA) level in the brain; the latter induces several deficiencies and disorders, including changes in neuronal cytomorphology and apoptosis. Therefore, we proposed that REMS- and REMSD-associated modulation of NA level might affect neuronal plasticity and affect brain functions. Male albino rats were REMS deprived by flower-pot method for 6 days, and its effects were compared with home cage and large platform controls as well as post-REMSD recovered and REMS-deprived prazosin (α1-adrenoceptor antagonist)-treated rats. We observed that REMSD reduced CA1 and CA3 neuronal dendritic length, branching, arborization, and spine density, while length of active zone and expressions of pre- as well as post-synaptic proteins were increased as compared to controls; interestingly, prazosin prevented most of the effects in vivo. Studies on primary culture of neurons from chick embryo brain confirmed that NA at lower concentration(s) induced neuronal branching and arborization, while higher doses were destructive. The findings support our contention that REMSD adversely affects neuronal plasticity, branching, and synaptic scaffold, which explain the underlying cytoarchitectural basis of REMSD-associated patho-physio-behavioral changes. Consolidation of findings of this study along with that of our previous reports suggest that the neuronal disintegration could be due to either withdrawal of direct protective and proliferative role of low dose of NA or indirect effect of high dose of NA or both.


Asunto(s)
Hipocampo/fisiopatología , Plasticidad Neuronal/fisiología , Norepinefrina/metabolismo , Privación de Sueño/fisiopatología , Animales , Embrión de Pollo , Masculino , Plasticidad Neuronal/efectos de los fármacos , Norepinefrina/farmacología , Ratas , Ratas Wistar
6.
Cell Mol Neurobiol ; 38(2): 441-457, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28353187

RESUMEN

Rapid eye movement sleep deprivation-associated elevated noradrenaline increases and decreases neuronal and glial Na-K ATPase activity, respectively. In this study, using C6 cell-line as a model, we investigated the possible intracellular molecular mechanism of noradrenaline-induced decreased glial Na-K ATPase activity. The cells were treated with noradrenaline in the presence or absence of adrenoceptor antagonists, modulators of extra- and intracellular Ca++ and modulators of intracellular signalling pathways. We observed that noradrenaline acting on ß-adrenoceptor decreased Na-K ATPase activity and mRNA expression of the catalytic α2-Na-K ATPase subunit in the C6 cells. Further, cAMP and protein kinase-A mediated release of intracellular Ca++ played a critical role in such decreased α2-Na-K ATPase expression. In contrast, noradrenaline acting on ß-adrenoceptor up-regulated the expression of regulatory ß2-Na-K ATPase subunit, which although was cAMP and Ca++ dependent, was independent of protein kinase-A and protein kinase-C. Combining these with previous findings (including ours) we have proposed a working model for noradrenaline-induced suppression of glial Na-K ATPase activity and alteration in its subunit expression. The findings help understanding noradrenaline-associated maintenance of brain excitability during health and altered states, particularly in relation to rapid eye movement sleep and its deprivation when the noradrenaline level is naturally altered.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Líquido Intracelular/enzimología , Receptores Adrenérgicos beta/fisiología , ATPasa Intercambiadora de Sodio-Potasio/biosíntesis , ATPasa Intercambiadora de Sodio-Potasio/genética , Animales , Carbazoles/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Líquido Intracelular/efectos de los fármacos , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Pirroles/farmacología , Ratas , Sodio/farmacología , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores
8.
Neurochem Res ; 40(8): 1747-57, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26148532

RESUMEN

Rapid eye movement sleep (REMS) modulates Na-K ATPase activity and maintains brain excitability. REMS deprivation (REMSD)-associated increased Na-K ATPase activity is mediated by noradrenaline (NA) acting on α1-adrenoceptor (AR) in the brain. It was shown that NA-induced increased Na-K ATPase activity was due to allosteric modulation as well as increased turnover of the enzyme. Although the former has been studied in detail, our understanding on the latter was lacking, which we have studied. Male Wistar rats were REMS deprived for 4-days by classical flower-pot method; suitable control experiments were conducted. In another set, α1-AR antagonist prazosin (PRZ) was i.p. injected 48 h REMSD onward. At the end of experiments rats were sacrificed by cervical dislocation and brains were removed. Synaptosomes prepared from the brains were used to estimate Na-K ATPase activity as well as protein expressions of different isoforms of the enzyme subunits using western blot. REMSD significantly increased synaptosomal Na-K ATPase activity and that was due to differential increase in the expressions of α1-, α2- and α3-isoforms, but not that of ß1- and ß2-isoforms. PRZ reduced the REMSD-induced increased Na-K ATPase activity and protein expressions. We also observed that the increased Na-K ATPase subunit expression was not due to enhanced mRNA synthesis, which suggests the possibility of post-transcriptional regulation. Thus, the findings suggest that REMSD-associated increased Na-K ATPase activity is due to elevated level of α-subunit of the enzyme and that is induced by NA acting on α1-AR mediated mRNA-stabilization.


Asunto(s)
Encéfalo/enzimología , Norepinefrina/farmacología , Subunidades de Proteína/biosíntesis , Receptores Adrenérgicos alfa 1/biosíntesis , Privación de Sueño/enzimología , Sueño REM/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Encéfalo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Masculino , Norepinefrina/fisiología , Ratas , Ratas Wistar , Sueño REM/efectos de los fármacos
9.
Neuropharmacology ; 247: 109861, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38331315

RESUMEN

Sleep is an instinct behavior, and its significance and functions are still an enigma. It is expressed throughout one's life and its loss affects psycho-somatic and physiological processes. We had proposed that it might maintain a fundamental property of the neurons and the brain. In that context, it was shown that sleep, rapid eye movement sleep (REMS) in particular, by regulating noradrenaline (NA), maintains the brain excitability. It was also reported that sleep-loss affected memory, reaction time and decision-making ability among others. However, as there was lack of clarity on the cause-and-effect relationship as to how the sleep-loss could affect these basic behaviors, their association was questioned and it was difficult to propose a cure or at least ways and means to ameliorate the symptoms. Also, we wanted to conduct the studies in a simpler model system so that conducting future molecular studies might be easier. Hence, using zebrafish as a model we evaluated if sleep-loss affected the basic decision-making ability, a cognitive process and if the effect was induced by NA. Indeed, our findings confirmed that upon sleep-deprivation, the cognitive decision-making ability of the prey zebrafish was compromised to protect itself by running away from the reach of the exposed predator Tiger Oscar (TO) fish. Also, we observed that upon sleep-loss the axonal arborization of the prey zebrafish brain was reduced. Interestingly, the effects were prevented by prazosin (PRZ), an α1-adrenoceptor (AR) antagonist and when the zebrafish recovered from the lost sleep.


Asunto(s)
Norepinefrina , Pez Cebra , Animales , Norepinefrina/farmacología , Privación de Sueño , Sueño , Neuronas , Receptores Adrenérgicos alfa 1/fisiología
10.
J Neurosci Res ; 91(11): 1450-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23996611

RESUMEN

Benzo[a]pyrene (B[a]P), a carcinogen, affects brain development, learning, and memory. Isolated studies have reported that B[a]P elevates noradrenaline (NA) level that may modulate neuronal growth, learning, and memory. Therefore, we investigated in vivo and in vitro the effects of B[a]P on learning and memory and its possible mechanism of action. Intracisternal administration of B[a]P on postnatal day 5 significantly reduced learning and memory in adolescent rats as observed by probe test using the Morris water maze. The density of both the subunits of the N-methyl-D-aspartate (NMDA) receptor, NMDAR1 and NMDAR2B, significantly increased in the hippocampus. In vitro, B[a]P significantly increased NMDAR1 in both C6 and Neuro2a cell lines, whereas NMDAR2B was significantly increased in C6 but was significantly decreased in Neuro2a. Pretreatment with NA prevented the B[a]P-induced effect on NMDAR1 expression in both cell lines. However, although NA prevented the B[a]P-mediated increase in NMDAR2B expression in C6, it further potentiated the decrease of NMDAR2B in Neuro2a cells. Also, NA prevented the B[a]P-induced increase in intracellular Ca(2+) both in C6 and in Neuro2a. Our findings show that postnatal exposure of developing rats to B[a]P impairs learning and memory even when the rats became adolescent. We also observed that the effects were mediated by elevated intracellular Ca(2+) levels and increased expression of NMDAR; furthermore, NA exerted a protective effect by modulating those factors. NA differentially affects neurons and glia, which may have a compensatory role during toxic insults, especially from B[a]P.


Asunto(s)
Benzo(a)pireno/toxicidad , Encéfalo/metabolismo , Carcinógenos/toxicidad , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Norepinefrina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Inmunohistoquímica , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar
11.
Physiol Behav ; 271: 114352, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37714322

RESUMEN

Rapid eye movement (REM) sleep plays a significant role in visuospatial learning and memory consolidation; however, its mechanism of action is unknown. Rapid eye movements (REMs), a characteristic active feature of REM sleep, is a potential correlate of neural processing for visual memory consolidation. The superior colliculus (SC) plays a central role in oculomotor control and spatial localization of objects in the visual field. We proposed that local reversible inactivation of the SC during post-learning sessions might interfere with REMs and negatively impact REM sleep associated consolidation of the visuospatial learnt task. Under gaseous anesthesia, bilateral cannulae aiming SC and electrodes for recording electrophysiological signals to classify sleep-waking were implanted. Following standard protocol, all rats were subjected to Morris water maze (MWM) training for 5 consecutive days followed by probe trial. After MWM training, on all except the probe test days, the rat SC were bilaterally infused with either vehicle (control, Group 1), Lidocaine hydrochloride a local anesthetic (Lox 2%, Group 2), or muscimol (Mus, GABA agonist, Group 3) and sleep-wakefulness recorded after day 1, 4, and post-probe learning sessions. Post-learning, compared to vehicle, Mus treated group significantly decreased REMs, phasic REM sleep, percent time spent in REM sleep and REM sleep frequency/hr. Also, during probe test, the escape latency was significantly increased, and the percentage time spent in the platform quadrant were significantly decreased in both, Mus and Lox 2% treated rats, while the number of platform location crossings was decreased in Mus treated group. The results showed that Lox 2% and Mus into SC reduced consolidation of visuospatial learning. The findings support our contention that SC mediated activation of REMs exerts a positive influence in processing and consolidation of visual learning during REM sleep. The findings explain the role of REMs during REM sleep in visual memory consolidation.

12.
Behav Brain Res ; 438: 114177, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36306944

RESUMEN

The superior colliculus (SC) is associated with visual attention, spatial navigation, decision making, escape and approach responses, some of which are important for defence and survival in rodents. SC helps in initiating and controlling saccadic eye movements and gaze during wakefulness. It is also activated during rapid eye movement (REM) sleep associated rapid eye movements (REMs). To investigate the contribution of SC in sleep-wake behaviour, we have demonstrated that manipulation of SC with scopolamine, carbachol, muscimol, picrotoxin and MK-801 decreased the amount of REM sleep. We observed that scopolamine and picrotoxin as well as muscimol decreased REM sleep frequency. MK-801 decreased percent amount of REM sleep, however, neither the frequency nor the duration/episode was affected. The cholinergic and GABA-ergic modulation of SC affecting REM sleep may be involved in REM sleep associated visuo-spatial learning and memory consolidation, which however, need to be confirmed. Furthermore, the results suggest involvement of efferent from SC in modulation of sleep-waking via the brainstem sleep regulating areas.


Asunto(s)
Sueño REM , Colículos Superiores , Ratas , Animales , Sueño REM/fisiología , Picrotoxina , Muscimol/farmacología , Maleato de Dizocilpina , Ácido gamma-Aminobutírico/fisiología , Vigilia/fisiología , Escopolamina/farmacología , Colinérgicos
13.
Sleep Med ; 110: 25-34, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37524037

RESUMEN

Rapid eye movement sleep (REMS) is essential for leading normal healthy living at least in higher-order mammals, including humans. In this review, we briefly survey the available literature for evidence linking cytomorphometric changes in the brain due to loss of REMS. As a mechanism of action, we add evidence that REMS loss elevates noradrenaline (NA) levels in the brain, which affects neuronal cytomorphology. These changes may be a compensatory mechanism as the changes return to normal after the subjects recover from the loss of REMS or if during REMS deprivation, the subjects are treated with NA-adrenoceptor antagonist prazosin (PRZ). We had proposed earlier that one of the fundamental functions of REMS is to maintain the level of NA in the brain. We elaborate on this idea to propose that if REMS loss continues without recovery, the sustained level of NA breaks down neurophysiologically active compensatory mechanism/s starting with changes in the neuronal cytomorphology, followed by their degeneration, leading to acute and chronic pathological conditions. Identification of neuronal cytomorphological changes could prove to be of significance for predicting future neuronal (brain) damage as well as an indicator for REMS health. Although current brain imaging techniques may not enable us to visualize changes in neuronal cytomorphology, given the rapid technological progress including use of artificial intelligence, we are optimistic that it may be a reality soon. Finally, we propose that maintenance of optimum REMS must be considered a criterion for leading a healthy life.


Asunto(s)
Inteligencia Artificial , Sueño REM , Animales , Humanos , Sueño REM/fisiología , Encéfalo/patología , Privación de Sueño/complicaciones , Prazosina , Mamíferos
14.
Neuropharmacology ; 237: 109621, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37276957

RESUMEN

The role of dopamine (DA)-ergic neurons in ventral tegmental area (VTA) in schizophrenia, depression, hallucinations have been extensively studied. Rapid eye movement sleep (REMS), the closest objective correlate of dream and hallucination, is disrupted during these psychological dysfunctions; however, it was unknown if there is any common neuronal substrate for their regulation. Interactions among locus coeruleus (LC) REM-OFF and pedunculopontine tegmentum (PPT) REM-ON neurons have been reported to regulate REMS in health and diseases. Recently we have reported that PPT neurons modulate VTA and REMS. However, although VTA-DA neurons receive projections from LC and PPT, their role in REMS regulation was unclear. We proposed that the LC and PPT might intermittently modulate VTA-DA neurons and modulate REMS. Male Wistar rats were surgically prepared and electrophysiological wakefulness-sleep-REMS recorded in chronic freely moving condition. We employed RNAi induced downregulation of tyrosine hydroxylase (TH) to evaluate the role of VTA-DA in regulating REMS. We observed that TH-knockdown in VTA decreased REMS in experimental rats, which returned to baseline upon PPT stimulation. Thus, VTA-DA neurons are activated by the REM-ON neurons to modulate REMS, the closest objectively recordable correlate of dreams. In these animals, LC stimulation altered Non-REMS and waking. Based on the findings we have discussed the role of VTA neurochemical circuitry in REMS regulation and their possible implications with REMS-associated dreaming and hallucination in health and diseases.


Asunto(s)
Locus Coeruleus , Sueño REM , Ratas , Masculino , Animales , Locus Coeruleus/fisiología , Sueño REM/fisiología , Dopamina/fisiología , Área Tegmental Ventral , Ratas Wistar , Neuronas/fisiología , Alucinaciones
15.
Brain Sci ; 14(1)2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38275513

RESUMEN

Wear and tear are natural processes for all living and non-living bodies. All living cells and organisms are metabolically active to generate energy for their routine needs, including for survival. In the process, the cells are exposed to oxidative load, metabolic waste, and bye-products. In an organ, the living non-neuronal cells divide and replenish the lost or damaged cells; however, as neuronal cells normally do not divide, they need special feature(s) for their protection, survival, and sustenance for normal functioning of the brain. The neurons grow and branch as axons and dendrites, which contribute to the formation of synapses with near and far neurons, the basic scaffold for complex brain functions. It is necessary that one or more basic and instinct physiological process(es) (functions) is likely to contribute to the protection of the neurons and maintenance of the synapses. It is known that rapid eye movement sleep (REMS), an autonomic instinct behavior, maintains brain functioning including learning and memory and its loss causes dysfunctions. In this review we correlate the role of REMS and its loss in synaptogenesis, memory consolidation, and neuronal degeneration. Further, as a mechanism of action, we will show that REMS maintains noradrenaline (NA) at a low level, which protects neurons from oxidative damage and maintains neuronal growth and synaptogenesis. However, upon REMS loss, the level of NA increases, which withdraws protection and causes apoptosis and loss of synapses and neurons. We propose that the latter possibly causes REMS loss associated neurodegenerative diseases and associated symptoms.

16.
Neuropharmacology ; 206: 108940, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34982973

RESUMEN

The interaction among the acetylcholine (ACh)-ergic REM-ON neurons in the pedunculo-pontine area (PPT), noradrenergic REM-OFF neurons in locus coeruleus (LC) and GABA-ergic neurons in the regulation of rapid eye movement sleep (REMS) have been studied in relative details; however, many questions including the role of dopamine (DA) remain unanswered. The ventral tegmental area (VTA) is rich in DA-ergic neurons, which have been implicated with schizophrenia and depression, when REMS is significantly affected. Also, some of the symptoms of REMS and these diseases are common. As the ACh-ergic REM-ON neurons in the PPT project to VTA, we proposed that such inputs might affect REMS, dreams and hallucinations. We recorded sleep-wake-REMS in freely moving, chronically prepared rats under three controlled experimental conditions. In different sets of experiments, either the ACh-ergic inputs to the VTA were blocked by local microinjection of Scopolamine (Scop) alone, or, the PPT neurons were bilaterally stimulated by Glutamate (Glut), or, the PPT neurons were stimulated by Glut in presence of Scop into the VTA. It was observed that Glut into PPT and Scop into the VTA significantly increased and decreased REMS, respectively. Additionally, PPT stimulation induced increased REMS was prevented in the presence of Scop into the VTA. Based on these findings we propose that inputs from ACh-ergic REM-ON neurons to VTA increase REMS and it could be a possible circuitry for expressions of hallucinations and dreams.


Asunto(s)
Neuronas Colinérgicas/fisiología , Neuronas Dopaminérgicas/fisiología , Tegmento Pontino/fisiología , Sueño REM/fisiología , Área Tegmental Ventral/fisiología , Animales , Antagonistas Colinérgicos/farmacología , Ratas , Escopolamina/farmacología , Sueño REM/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos
17.
Brain Sci ; 12(12)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36552184

RESUMEN

Immune function and sleep are two normal physiological processes to protect the living organism from falling sick. There is hardly any disease in which they remain unaffected, though the quantum of effect may differ. Therefore, we propose the existence of a strong correlation between sleep (quality or quantity) and immune response. This may be supported by the fact that sleep loss modulates many of the immunological molecules, which includes interferons; however, not much is known about their mechanism of action. Sleep is divided into rapid eye movement sleep (REMS) and non-REMS. For practical reasons, experimental studies have been conducted mostly by inducing loss of REMS. It has been shown that withdrawal of noradrenaline (NA) is a necessity for generation of REMS. Moreover, NA level increases in the brain upon REMS loss and the elevated NA is responsible for many of the sleep loss-associated symptoms. In this review, we describe how sleep (and its disturbance/loss) modulates the immune system by modulating the NA level in the brain or vice versa to maintain immune functions, physiological homeostasis, and normal healthy living. The increased levels of NA during REMS loss may cause neuroinflammation possibly by glial activation (as NA is a key modulator of microglia). Therefore, maintaining sleep hygiene plays a crucial role for a normal healthy living.

18.
Neuropharmacology ; 193: 108607, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34023337

RESUMEN

Dreams appear intermittently during phasic rapid eye movement sleep (REMS). Although reasonable progress has been made about neuro-physio-pharmacological mechanism of appearance of REMS, appearance of dreams is a mystery. Isolated studies have reported that substantia nigra (SN) withdraws inhibition from pedunculo-pontine tegmentum (PPT) acetylcholine (ACh)-ergic REM-ON neurons to trigger REMS; some REM-ON neurons become phasically active during REMS; amygdala (Amyg), a limbic structure associated with emotions, may be related with dreaming like state; Amyg receives projections from both SN-Dopamine (DA)-ergic and PPT-ACh-ergic neurons. Collating these isolated findings, we proposed that on the background of REMS, SN-DA-ergic and PPT-ACh-ergic inputs phasically activate Amyg-neurons to manifest dreams. In the absence of better criteria, we recorded electrophysiological characteristics of REMS as the closest objective read-out for dreams in surgically prepared, chronic, freely moving rats. Microinjection of either DA-ergic or ACh-ergic agonist [Quinpirole (Qnp) or Carbachol (Carb)] bilaterally into Amyg increased, while antagonists [Haloperidol (Hal) or Scopolamine (Scop)] reduced REMS. Electrical stimulation of either bilateral SN or PPT increased REMS, which however, was prevented when stimulated in presence of Hal or Scop, respectively into the Amyg. These findings confirm and support our contention that SN-DA-ergic and PPT-ACh-ergic inputs integrate in Amyg for REMS regulation. Further, subject to confirmation in humans, we propose that on the background of REMS, some phasic PPT-ACh-ergic-REM-ON neurons intermittently trigger some neurons in Amyg, the area known to be associated with memory and emotions, causing intermittent appearance of REMS-associated dreams and in REMS behavior disorder.


Asunto(s)
Amígdala del Cerebelo/fisiología , Neuronas Dopaminérgicas/fisiología , Tegmento Pontino/fisiología , Sueño REM/fisiología , Sustancia Negra/fisiología , Vigilia/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Carbacol/farmacología , Neuronas Colinérgicas , Estimulación Eléctrica , Haloperidol/farmacología , Masculino , Tegmento Pontino/efectos de los fármacos , Quinpirol/farmacología , Ratas , Ratas Wistar , Escopolamina/farmacología , Sueño REM/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Vigilia/efectos de los fármacos
19.
Neurosci Lett ; 745: 135631, 2021 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-33444674

RESUMEN

Flowerpot method of rapid eye movement sleep (REMS) deprivation (REMSD) has been most extensively used in experiments to decipher the functions of REMS. The most common but serious criticism of this method has been presumed stress experienced by the experimental animals. The lack of systematic studies with appropriate controls to resolve this issue prompted this study. We have compared serum corticosterone levels as a marker of stress in male rats under REMSD by the flowerpot method and multiple types of control conditions. Additionally, to maintain consistency and uniformity of REMSD among groups, in the same rats, we estimated brain Na-K ATPase activity, which has been consistently reported to increase upon REMSD. The most effective method was one rat in single- or multiple-platforms set-up in a pool because it significantly increased Na-K ATPase activity without elevating serum corticosterone level. More than one rat in multiple platform set-up was ineffective and must be avoided. Also, large platform- and recovery-controls must be carried out simultaneously to rule out non-specific confounding effects.


Asunto(s)
Encéfalo/metabolismo , Corticosterona/sangre , Privación de Sueño/sangre , Sueño REM/fisiología , Estrés Psicológico/sangre , Animales , Biomarcadores/sangre , Masculino , Ratas , Ratas Wistar , Privación de Sueño/psicología , Estrés Psicológico/psicología
20.
Mol Cell Biochem ; 336(1-2): 3-16, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19823772

RESUMEN

Rapid eye movement (REM) sleep is a unique phenomenon expressed in all higher forms of animals. Its quantity varies in different species and with ageing; it is also affected in several psycho-somatic disorders. Several lines of studies showed that after REM sleep loss, the levels of noradrenaline (NA) increase in the brain. The NA in the brain modulates neuronal Na-K ATPase activity, which helps maintaining the brain excitability status. The detailed mechanism of increase in NA level after REM sleep loss and the effect of NA on stimulation of Na-K ATPase in the neurons have been discussed. The findings have been reviewed and discussed with an aim to understand the role of REM sleep in maintaining brain excitability status.


Asunto(s)
Encéfalo/fisiopatología , Neuronas/fisiología , Norepinefrina/fisiología , Privación de Sueño/fisiopatología , Sueño REM/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Encéfalo/metabolismo , Señalización del Calcio , Membrana Celular/metabolismo , Activación Enzimática , Potenciales Evocados Motores/fisiología , Humanos , Isoenzimas , Norepinefrina/análisis , Receptores Adrenérgicos/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA