Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
J Neurosci ; 44(28)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38830763

RESUMO

Chronic sleep disruption (CSD), from insufficient or fragmented sleep and is an important risk factor for Alzheimer's disease (AD). Underlying mechanisms are not understood. CSD in mice results in degeneration of locus ceruleus neurons (LCn) and CA1 hippocampal neurons and increases hippocampal amyloid-ß42 (Aß42), entorhinal cortex (EC) tau phosphorylation (p-tau), and glial reactivity. LCn injury is increasingly implicated in AD pathogenesis. CSD increases NE turnover in LCn, and LCn norepinephrine (NE) metabolism activates asparagine endopeptidase (AEP), an enzyme known to cleave amyloid precursor protein (APP) and tau into neurotoxic fragments. We hypothesized that CSD would activate LCn AEP in an NE-dependent manner to induce LCn and hippocampal injury. Here, we studied LCn, hippocampal, and EC responses to CSD in mice deficient in NE [dopamine ß-hydroxylase (Dbh)-/-] and control male and female mice, using a model of chronic fragmentation of sleep (CFS). Sleep was equally fragmented in Dbh -/- and control male and female mice, yet only Dbh -/- mice conferred resistance to CFS loss of LCn, LCn p-tau, and LCn AEP upregulation and activation as evidenced by an increase in AEP-cleaved APP and tau fragments. Absence of NE also prevented a CFS increase in hippocampal AEP-APP and Aß42 but did not prevent CFS-increased AEP-tau and p-tau in the EC. Collectively, this work demonstrates AEP activation by CFS, establishes key roles for NE in both CFS degeneration of LCn neurons and CFS promotion of forebrain Aß accumulation, and, thereby, identifies a key molecular link between CSD and specific AD neural injuries.


Assuntos
Peptídeos beta-Amiloides , Cisteína Endopeptidases , Hipocampo , Locus Cerúleo , Norepinefrina , Privação do Sono , Animais , Peptídeos beta-Amiloides/metabolismo , Norepinefrina/metabolismo , Camundongos , Hipocampo/metabolismo , Hipocampo/patologia , Privação do Sono/metabolismo , Privação do Sono/patologia , Masculino , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/genética , Fragmentos de Peptídeos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dopamina beta-Hidroxilase/metabolismo , Dopamina beta-Hidroxilase/genética , Proteínas tau/metabolismo , Feminino , Degeneração Neural/patologia , Degeneração Neural/metabolismo , Degeneração Neural/genética
2.
Sci Transl Med ; 15(693): eade6285, 2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37099634

RESUMO

Sleep loss is associated with cognitive decline in the aging population and is a risk factor for Alzheimer's disease (AD). Considering the crucial role of immunomodulating genes such as that encoding the triggering receptor expressed on myeloid cells type 2 (TREM2) in removing pathogenic amyloid-ß (Aß) plaques and regulating neurodegeneration in the brain, our aim was to investigate whether and how sleep loss influences microglial function in mice. We chronically sleep-deprived wild-type mice and the 5xFAD mouse model of cerebral amyloidosis, expressing either the humanized TREM2 common variant, the loss-of-function R47H AD-associated risk variant, or without TREM2 expression. Sleep deprivation not only enhanced TREM2-dependent Aß plaque deposition compared with 5xFAD mice with normal sleeping patterns but also induced microglial reactivity that was independent of the presence of parenchymal Aß plaques. We investigated lysosomal morphology using transmission electron microscopy and found abnormalities particularly in mice without Aß plaques and also observed lysosomal maturation impairments in a TREM2-dependent manner in both microglia and neurons, suggesting that changes in sleep modified neuro-immune cross-talk. Unbiased transcriptome and proteome profiling provided mechanistic insights into functional pathways triggered by sleep deprivation that were unique to TREM2 and Aß pathology and that converged on metabolic dyshomeostasis. Our findings highlight that sleep deprivation directly affects microglial reactivity, for which TREM2 is required, by altering the metabolic ability to cope with the energy demands of prolonged wakefulness, leading to further Aß deposition, and underlines the importance of sleep modulation as a promising future therapeutic approach.


Assuntos
Doença de Alzheimer , Amiloidose , Camundongos , Animais , Microglia/metabolismo , Privação do Sono/complicações , Privação do Sono/metabolismo , Privação do Sono/patologia , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Placa Amiloide/patologia , Modelos Animais de Doenças , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo
3.
Shanghai Kou Qiang Yi Xue ; 31(2): 148-155, 2022 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-36110071

RESUMO

PURPOSE: The aim of this study was to investigate the morphological changes of condylar cartilage of temporomandibular joint (TMJ) and the expression changes of IL-1ß,TNF-α,IGF-1 and VEGF in condylar cartilage of TMJ by establishing a chronic sleep deprivation model in rats. METHODS: Sixty rats were randomly divided into experimental group, control group and recovery group. Modified multiple platforms method (MMPM) was used to build chronic sleep deprivation models in experimental and recovery groups. Rats in the recovery group received 1 week of cage feeding after sleep deprivation. H-E staining was used to observe morphological change of the condyle. Immunohistochemical method was performed to detect the changes of IL-1ß, TNF-α, IGF-1 and VEGF. The data was processed by using SPSS 23.0 software package. RESULTS: MMPM can establish chronic sleep deprivation model effectively. H-E staining showed condylar cartilage of the experimental group was split stripped, and the boundaries of cartilage cell layer became blurred. Compared with the control group, the recovery group had less cracks in the fibrous layer or some of the cracks were occupied by fibrous tissue. Immunohistochemistry showed that the positive expression intensity of IL-1ß and TNF-α in the experimental group was significantly higher than in the control group (P<0.05), the positive expression intensity in the recovery group was significantly lower than in the experimental group(P<0.05). The positive expression intensity of IGF-1 and VEGF in the experimental group was significantly higher than in the control group(P<0.05). The expression of IGF-1 and VEGF decreased significantly in the recovery group which received sleep deprivation no more than 3 weeks(P<0.05). CONCLUSIONS: Chronic sleep deprivation can increase the expression of IL-1ß, TNF-α and VEGF in condylar cartilage and aggravate osteoarthritis. Chronic sleep deprivation can lead to increase of IGF-1 in condylar cartilage tissue, which plays a crucial role in protecting and promoting the reconstruction of condylar cartilage. After chronic sleep deprivation, the expressions of IL-1ß, TNF-α, IGF-1 and VEGF in the condylar cartilage of rats were decreased after 1 week of recovery, and the condylar cartilage underwent restorative reconstruction.


Assuntos
Cartilagem , Animais , Cartilagem/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Côndilo Mandibular/metabolismo , Ratos , Privação do Sono/patologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Biomed Pharmacother ; 148: 112765, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35247715

RESUMO

Oxidative stress has been linked with sleep deprivation (SD)-induced pathological conditions and reproductive dysfunction. On the other hand, glutamine has been established to have antioxidant property. However, the impact of SD, with or without glutamine, on male reproductive function is yet to be elucidated. Thus, this study was designed to investigate the role of SD, with or without glutamine, on male reproductive function and possible associated mechanisms. Ten-week old male Wistar rats weighing 175.6 g± 0.42 were randomly assigned into vehicle that received per os (p.o.) distilled water, glutamine (1 g/kg; po), SD, and SD + glutamine that received treatments as glutamine and SD. Treatment/exposure lasted for 72 h. The results showed that SD led to reduced body weight, seminiferous luminal and epididymal sperm density, low sperm quality, increased testicular and epididymal malondialdehyde, uric acid, DNA fragmentation, and testicular injury markers. In addition, SD caused a reduction in reduced glutathione level and activities of superoxide dismutase, catalase, glucose-6-phosphate dehydrogenase, glutathione peroxidase, and glutathione-S-transferase. Also, SD increased tumor necrotic factor-α, interleukin-1ß, and nuclear factor-kappa B levels. Furthermore SD led to impaired libido and erectile dysfunction, and suppression of circulatory nitric oxide, gonadotropins and testosterone, and penile cGMP. However, glutamine attenuated the effects induced by SD. Taken together, the findings of this study demonstrate that SD induces reproductive dysfunction via glutathione-dependent defense depletion and down-regulation of NO/cGMP signaling, which was abolished by glutamine supplementation.


Assuntos
GMP Cíclico/metabolismo , Glutamina/farmacologia , Óxido Nítrico/metabolismo , Disfunções Sexuais Fisiológicas/patologia , Privação do Sono/patologia , Testículo/patologia , Animais , Antioxidantes/farmacologia , Epididimo/efeitos dos fármacos , Epididimo/metabolismo , Disfunção Erétil/patologia , Libido/efeitos dos fármacos , Libido/fisiologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Distribuição Aleatória , Ratos , Ratos Wistar , Testículo/efeitos dos fármacos
5.
Cell ; 184(5): 1348-1361.e22, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636128

RESUMO

Clonal hematopoiesis, a condition in which individual hematopoietic stem cell clones generate a disproportionate fraction of blood leukocytes, correlates with higher risk for cardiovascular disease. The mechanisms behind this association are incompletely understood. Here, we show that hematopoietic stem cell division rates are increased in mice and humans with atherosclerosis. Mathematical analysis demonstrates that increased stem cell proliferation expedites somatic evolution and expansion of clones with driver mutations. The experimentally determined division rate elevation in atherosclerosis patients is sufficient to produce a 3.5-fold increased risk of clonal hematopoiesis by age 70. We confirm the accuracy of our theoretical framework in mouse models of atherosclerosis and sleep fragmentation by showing that expansion of competitively transplanted Tet2-/- cells is accelerated under conditions of chronically elevated hematopoietic activity. Hence, increased hematopoietic stem cell proliferation is an important factor contributing to the association between cardiovascular disease and clonal hematopoiesis.


Assuntos
Aterosclerose/patologia , Hematopoiese Clonal , Células-Tronco Hematopoéticas/patologia , Envelhecimento/patologia , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Medula Óssea/metabolismo , Proliferação de Células , Evolução Clonal , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Privação do Sono/patologia
6.
Biochem Biophys Res Commun ; 534: 632-638, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33213844

RESUMO

Caffeine is a common stimulant widely existed in food and has stimulatory effects on the central nervous system, shift-work individuals often rely on caffeine to maintain attention and keep awake. Although sleep deprivation (SD) is widely considered as an independent risk factor for cognition retardations, however, little is well understood about the synergistic role of caffeine dosage and SD for cognitive performance. This research intended to investigate the underlying molecular mechanism of varying caffeine doses on cognitive function after sleep deprivation. The results revealed that SD attenuated the cognitive dysfunction, associated with ultrastructure damage and pyramidal neuron loss in the hippocampus, decreased in the level of VIP and AVP. SD also significantly accelerated the neuropeptide-associated apoptosis in the hippocampus, which may modulate via the cAMP-PKA-CREB signal path axis and activation of the downstream apoptosis genes. Additionally, the data indicated that low-dose caffeine (LC) contributed to cognitive enhancement, and high-dose caffeine (HC) aggravated cognitive impairment by modulating hippocampal neuronal apoptosis. Our studies suggest that caffeine, particularly in high dosage, may be a potential factor to influence the neurocognitive outcome caused by sleep loss, and the appropriate amount of caffeine ingested after sleep deprivation deserves serious consideration.


Assuntos
Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Cognição/efeitos dos fármacos , Privação do Sono/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Arginina Vasopressina/metabolismo , Cafeína/administração & dosagem , Estimulantes do Sistema Nervoso Central/administração & dosagem , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/patologia , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Atividade Motora/efeitos dos fármacos , Nootrópicos/administração & dosagem , Nootrópicos/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Células Piramidais/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Privação do Sono/patologia , Privação do Sono/psicologia , Aprendizagem Espacial/efeitos dos fármacos , Peptídeo Intestinal Vasoativo/metabolismo
7.
Int J Mol Sci ; 21(22)2020 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-33233617

RESUMO

Obstructive sleep apnoea (OSA) is a prevalent disorder associated with increased cardiovascular, metabolic and neurocognitive morbidity. Recently, an increasing number of basic, clinical and epidemiological reports have suggested that OSA may also increase the risk of cancer, and adversely impact cancer progression and outcomes. This hypothesis is convincingly supported by biological evidence linking certain solid tumours and hypoxia, as well as by experimental studies involving cell and animal models testing the effects of intermittent hypoxia and sleep fragmentation that characterize OSA. However, the clinical and epidemiological studies do not conclusively confirm that OSA adversely affects cancer, even if they hold true for specific cancers such as melanoma. It is likely that the inconclusive studies reflect that they were not specifically designed to test the hypothesis or because of the heterogeneity of the relationship of OSA with different cancer types or even sub-types. This review critically focusses on the extant basic, clinical, and epidemiological evidence while formulating proposed directions on how the field may move forward.


Assuntos
Envelhecimento/genética , Hipóxia/genética , Neoplasias/genética , Obesidade/genética , Apneia Obstrutiva do Sono/genética , Privação do Sono/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Estudos de Coortes , Estudos Transversais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Obesidade/metabolismo , Obesidade/patologia , Fatores de Risco , Apneia Obstrutiva do Sono/metabolismo , Apneia Obstrutiva do Sono/patologia , Privação do Sono/metabolismo , Privação do Sono/patologia
8.
J Alzheimers Dis ; 74(3): 759-773, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32083588

RESUMO

Alzheimer's disease (AD) is the most common age-associated neurodegenerative disease featured by progressive learning and memory deficit, and Aß was identified as playing a key role in the process of AD and was theorized to be caused by the imbalance of production and clearance. Increasing evidence suggested an association between sleep deprivation and AD. Our recent study found that chronic sleep restriction (CSR) caused cognitive impairment and Aß accumulation in rats, but the underlining mechanism was unclear. In the present study, we investigated the effects of inflammation on Aß accumulation induced by CSR. We found that CSR significantly increased the expression of interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), and nitric oxide (NO) in brain, and the inflammatory factors levels were positively correlated with Aß42 deposition. Additionally, the inflammatory factors were correlated with BACE1, LRP-1, and RAGE levels in both the hippocampus and the prefrontal cortex. Furthermore, the plasma levels of IL-1ß, TNF-α, and NO were elevated after CSR, and the concentration of plasma inflammatory mediators were correlated with plasma levels of sLRP1 and sRAGE. These results suggested that the inflammation in brain and plasma might be involved in the CSR-induced Aß accumulation.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encefalite/metabolismo , Encefalite/patologia , Privação do Sono/metabolismo , Privação do Sono/patologia , Animais , Citocinas/metabolismo , Hipocampo/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Óxido Nítrico/metabolismo , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
9.
Biochem Biophys Res Commun ; 522(3): 749-756, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31787230

RESUMO

Sleep deprivation (SD) has detrimental effects on the physiological function of the brain. However, the underlying mechanism remains elusive. In the present study, we investigated the expression of candidate plasticity-related gene 15 (cpg15), a neurotrophic gene, and its potential role in SD using a REM-SD mouse model. Immunofluorescent and Western blot analysis revealed that the expression of cpg15 protein decreased in the hippocampus, ventral group of the dorsal thalamus (VENT), and somatosensory area of cerebral cortex (SSP) after 24-72 h of REM-SD, and the oxidative stress in these brain regions was increased in parallel, as indicated by the ratio of glutathione (GSH) to its oxidative product (GSSG). Over-expression of cpg15 in thalamus, hippocampus, and cerebral cortex mediated by AAV reduced the oxidative stress in these regions, indicating that the decrease of cpg15 might be a cause that augments oxidative stress in the sleep deprived mouse brain. Collectively, the results imply that cpg15 may play a protective function in the SD-subjected mouse brain via an anti-oxidative function. To our knowledge, this is the first time to provide evidences in the role of cpg15 against SD-induced oxidative stress in the brain.


Assuntos
Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo , Privação do Sono/metabolismo , Animais , Encéfalo/patologia , Células COS , Chlorocebus aethiops , Proteínas Ligadas por GPI/metabolismo , Glutationa/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Privação do Sono/patologia
10.
Biol. Res ; 53: 31, 2020. graf
Artigo em Inglês | LILACS | ID: biblio-1124214

RESUMO

BACKGROUND: In modern societies, sleep deprivation is a serious health problem. This problem could be induced by a variety of reasons, including lifestyle habits or neurological disorders. Chronic sleep deprivation (CSD) could have complex biological consequences, such as changes in neural autonomic control, increased oxidative stress, and inflammatory responses. The superior cervical ganglion (SCG) is an important sympathetic component of the autonomic nervous system. CSD can lead to a wide range of neurological consequences in SCG, which mainly supply innervations to circadian system and other structures. As the active component of Curcuma longa, curcumin possesses many therapeutic properties; including neuroprotective. This study aimed to evaluate the effect of CSD on the SCG histomorphometrical changes and the protective effect of curcumin in preventing these changes. METHODS: Thirty-six male rats were randomly assigned to the control, curcumin, CSD, CSD + curcumin, grid floor control, and grid floor + curcumin groups. The CSD was induced by a modified multiple platform apparatus for 21 days and animals were sacrificed at the end of CSD or treatment, and their SCGs removed for stereological and TUNEL evaluations and also spatial arrangement of neurons in this structure. RESULTS: Concerning stereological findings, CSD significantly reduced the volume of SCG and its total number of neurons and satellite glial cells in comparison with the control animals ( P < 0.05). Treatment of CSD with curcumin prevented these decreases. Furthermore, TUNEL evaluation showed significant apoptosis in the SCG cells in the CSD group, and treatment with curcumin significantly decreased this apoptosis ( P < 0.01). This decrease in apoptosis was observed in all control groups that received curcumin. CSD also changed the spatial arrangement of ganglionic neurons into a random pattern, whereas treatment with curcumin preserved its regular pattern. CONCLUSIONS: CSD could potentially induce neuronal loss and structural changes including random spatial distribution in the SCG neurons. Deleterious effects of sleep deprivation could be prevented by the oral administration of curcumin. Furthermore, the consumption of curcumin in a healthy person might lead to a reduction of cell death.


Assuntos
Animais , Masculino , Ratos , Privação do Sono/patologia , Privação do Sono/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Gânglio Cervical Superior/efeitos dos fármacos , Curcumina/farmacologia , Ratos Sprague-Dawley
11.
Biochem Biophys Res Commun ; 513(2): 452-459, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-30967262

RESUMO

Sleep deprivation affects the pathophysiology of immune-inflammatory skin diseases such as psoriasis. Increasing efforts are directed toward exploring potential mechanisms involving sleep deprivation and immune responses. However, studies focusing on the effects of different timing of sleep deprivation on skin inflammation are lacking. This study thus investigated the immunomodulatory effects of paradoxical sleep (PS) deprivation at different timing of psoriasiform process on skin inflammation in the psoriasis-like mouse model. Male adult C57BL/6 mice were divided into 5 groups (each n = 5): control group (CON), IMQ-S2 group [treating with 31.25 mg imiquimod (IMQ) per day for 5 days with 48 h of PS deprivation on the fourth and fifth day], S2-IMQ group (treating with 31.25 mg IMQ per day for 5 days with 48 h of PS deprivation on the first and second day), 31.25 group (treating with 31.25 mg IMQ per day for 5 days) and 62.5 group (treating with 62.5 mg IMQ per day for 5 days). Compared with IMQ-S2 group and 31.25 group, S2-IMQ group mice had significant increase of IL-17A mRNA level in skin lesions and lymph nodes, and more severe cutaneous inflammation. However, IMQ-S2 group had the highest IL-1ß mRNA level in skin lesions and the highest IL-6 mRNA level in lymph nodes among the three groups. Results of flow cytometry showed that frequencies of γδT cell, IL-17A+γδT cell, dendritic cell (DC) and MHCⅡ+ DC in lymph nodes of S2-IMQ group were significantly higher than IMQ-S2 group and 31.25 group, so was the frequency of γδT cells in skin lesions. However, the frequency of DCs in skin lesions of S2-IMQ group was significantly lower than IMQ-S2 group. These data suggest that PS deprivation at the early stage rather than the late stage of psoriasiform process exacerbates the skin inflammation through modulation of the immune system, which may involve that IL-1ß and IL-6 stimulated by PS deprivation in turn increasing IL-17 expression through activation and proliferation of γδT cells and DCs migration.


Assuntos
Inflamação/complicações , Psoríase/complicações , Pele/patologia , Privação do Sono/complicações , Animais , Células Dendríticas/imunologia , Células Dendríticas/patologia , Modelos Animais de Doenças , Inflamação/imunologia , Inflamação/patologia , Interleucina-17/imunologia , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Masculino , Camundongos Endogâmicos C57BL , Psoríase/imunologia , Psoríase/patologia , Pele/imunologia , Privação do Sono/imunologia , Privação do Sono/patologia
12.
Neuroimmunomodulation ; 25(2): 59-67, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30007965

RESUMO

OBJECTIVES: Sleep regulates immune function reciprocally and can affect the parameters that are directly involved in the immune response. Sleep deprivation is considered to be a stress-causing factor and is associated with impaired immune activity. It causes increased glucocorticoid concentrations by activating the hypothalamic-pituitary-adrenal axis; this can lead to a series of disorders that are associated with the prolonged or increased secretion of these hormones. The aim of this study was to evaluate the effects of sleep restriction (SR) on the development of pulmonary experimental metastasis and the modulation of the tumor immune response. METHODS: The SR protocol was accomplished by depriving C57BL/6 male mice of sleep for 18 h/day for 2, 7, 14, and 21 days. The modified multiple-platforms method was used for SR. RESULTS: The results showed that cytotoxic cells (i.e., natural killer [NK] and CD8+ T cells) were reduced in number and regulatory T cells were predominant in the tumor microenvironment. Sleep-restricted mice also exhibited a reduced number of dendritic cells in their lymph nodes, which may have contributed to the ineffective activation of tumor-specific T cells. Peripheral CD4+ and CD8+ T cells were also reduced in the sleep-restricted mice, thus indicating an immunosuppressive status. CONCLUSIONS: Sleep dep-rivation induces failure in the activity of cells that are im-portant to the tumor immune response, both in the tumor microenvironment and on the periphery. This leads to the early onset and increased growth rate of lung metastasis.


Assuntos
Imunidade Celular/imunologia , Neoplasias Pulmonares/imunologia , Linfócitos/imunologia , Privação do Sono/imunologia , Microambiente Tumoral/imunologia , Animais , Neoplasias Pulmonares/patologia , Linfócitos/patologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Privação do Sono/patologia
13.
Neuropharmacology ; 139: 52-60, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29928886

RESUMO

Painful conditions and sleep disturbances are major public health problems worldwide and one directly affects the other. Sleep loss increases pain prevalence and severity; while pain disturbs sleep. However, the underlying mechanisms are largely unknown. Here we asked whether chronic sleep restriction for 6 h daily progressively increases pain sensitivity and if this increase is reversed after two days of free sleep. Also, whether the pronociceptive effect of chronic sleep restriction depends on the periaqueductal grey and on the nucleus accumbens, two key regions involved in the modulation of pain and sleep-wake cycle. We showed that sleep restriction induces a pronociceptive effect characterized by a significant decrease in the mechanical paw withdrawal threshold in rats. Such effect increases progressively from day 3 to day 12 remaining stable thereafter until day 26. Two consecutive days of free sleep were not enough to reverse the effect, not even to attenuate it. This pronociceptive effect depends on the periaqueductal grey and on the nucleus accumbens, since it was prevented by their excitotoxic lesion. Complementarily, chronic sleep restriction significantly increased c-Fos protein expression within the periaqueductal grey and the nucleus accumbens and this correlates with the intensity of the pronociceptive effect, suggesting that the greater the neural activity in this regions, the greater the effect. These findings may contribute not only to understand why painful conditions are more prevalent and severe among people who sleep poorly, but also to develop therapeutic strategies to prevent this, increasing the effectiveness of pain management in this population.


Assuntos
Núcleo Accumbens/fisiopatologia , Percepção da Dor/fisiologia , Limiar da Dor/fisiologia , Substância Cinzenta Periaquedutal/fisiopatologia , Privação do Sono/fisiopatologia , Animais , Masculino , N-Metilaspartato/toxicidade , Dor Nociceptiva/patologia , Dor Nociceptiva/fisiopatologia , Núcleo Accumbens/patologia , Substância Cinzenta Periaquedutal/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar , Privação do Sono/patologia , Fatores de Tempo , Tato
14.
Int J Mol Sci ; 19(6)2018 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-29843406

RESUMO

It is recognized that the properties of liquid water can be markedly different from those of bulk one when it is in contact with hydrophobic surfaces or is confined in nano-environments. Because our knowledge regarding water structure on the molecular level of dynamic equilibrium within a picosecond time scale is far from completeness all of water's conventionally known properties are based on inert "bulk liquid water" with a tetrahedral hydrogen-bonded structure. Actually, the strength of water's hydrogen bonds (HBs) decides its properties and activities. In this review, an innovative idea on preparation of metastable plasmon-activated water (PAW) with intrinsically reduced HBs, by letting deionized (DI) water flow through gold-supported nanoparticles (AuNPs) under resonant illumination at room temperature, is reported. Compared to DI water, the created stable PAW can scavenge free hydroxyl and 2,2-diphenyl-1-picrylhydrazyl radicals and effectively reduce NO release from lipopolysaccharide-induced inflammatory cells. Moreover, PAW can dramatically induce a major antioxidative Nrf2 gene in human gingival fibroblasts. This further confirms its cellular antioxidative and anti-inflammatory properties. In addition, innovatively therapeutic strategy of daily drinking PAW on inflammatory-related diseases based on animal disease models is demonstrated, examples being chronic kidney disease (CKD), chronic sleep deprivation (CSD), and lung cancer.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Ouro/química , Nanopartículas/química , Ressonância de Plasmônio de Superfície , Água/farmacologia , Animais , Anti-Inflamatórios/química , Antioxidantes/química , Compostos de Bifenilo/antagonistas & inibidores , Compostos de Bifenilo/química , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Ligação de Hidrogênio , Lipopolissacarídeos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Picratos/antagonistas & inibidores , Picratos/química , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Privação do Sono/tratamento farmacológico , Privação do Sono/metabolismo , Privação do Sono/patologia , Água/química
15.
FASEB J ; 32(10): 5390-5404, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29702026

RESUMO

Previous evidence has suggested that dietary supplementation with a bioactive dietary polyphenol preparation (BDPP) rescues impairment of hippocampus-dependent memory in a mouse model of sleep deprivation (SD). In the current study, we extend our previous evidence and demonstrate that a mechanism by which dietary BDPP protects against SD-mediated cognitive impairment is via mechanisms that involve phosphorylation of the mammalian target of rapamycin complex 1 and its direct downstream targets, including the eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) and the ribosomal protein S6 kinase ß-1 (p70S6K). In additional mechanistic studies in vitro, we identified the brain bioavailable phenolic metabolites derived from the metabolism of dietary BDPP that are responsible for the attenuation of SD-mediated memory impairments. On the basis of high-throughput bioavailability studies of brain bioavailable metabolites after dietary BDPP treatment, we found that select polyphenol metabolites [ e.g., cyanidin-3'- O-glucoside and 3-(3'-hydroxyphenyl) propionic acid] were able to rescue mTOR and p70S6K phosphorylation in primary cortico-hippocampal neuronal cultures, as well as rescue 4E-BP1 phosphorylation in response to treatment with 4EGI-1, a specific inhibitor of eIF4E-eIF4G interaction. Our findings reveal a previously unknown role for dietary polyphenols in the rescue of SD-mediated memory impairments via mechanisms involving the promotion of protein translation.-Frolinger, T., Smith, C., Cobo, C. F., Sims, S., Brathwaite, J., de Boer, S., Huang, J., Pasinetti, G. M. Dietary polyphenols promote resilience against sleep deprivation-induced cognitive impairment by activating protein translation.


Assuntos
Disfunção Cognitiva , Hipocampo , Neurônios , Polifenóis/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Privação do Sono , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Fator de Iniciação 4E em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Fosfoproteínas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Privação do Sono/tratamento farmacológico , Privação do Sono/metabolismo , Privação do Sono/patologia
16.
Mol Cell Biochem ; 449(1-2): 63-72, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29549603

RESUMO

Chronic sleep loss/fragmentation prevalent in the current 24/7 society is associated with irreversible consequences on health and overall wellbeing. Various studies have well documented the ill effects of acute sleep loss on cognitive functions of individuals; however, the underlying mechanism behind the chronic sleep loss is yet to be explored. The present study was aimed to investigate whether chronic sleep deprivation (CSD) triggers anxiety-like behaviour and memory decline in male Wistar rats. Rats were sleep deprived by placing them over slowly rotating drum (2 rpm) for 18 h (between 4 pm and 10 am) followed by 6 h of recovery sleep for 21 consecutive days. Post CSD regimen, rats were subjected to behavioural tests such as elevated plus maze (EPM), Novel Object Recognition (NOR) and Rotarod performance test and then sacrificed to remove brain for further molecular studies. The study demonstrated that CSD rats showed anxiogenic behaviour along with recognition memory decline compared to control rats. CSD rats further showed elevated levels of inflammatory cytokines (TNFα, IL-1ß) along with activation of NFκB and AP1 transcription factors in hippocampus and piriform cortex (PC) regions of brain. These observations were also accompanied by enhanced expression of GFAP and Iba1 in the two brain regions. The data suggest that CSD triggered low-grade neuroinflammation which caused anxiogenic response and recognition memory impairment. The study provides preliminary leads to further explore the role of astrocytes/microglial cells and inflammatory cytokines in mediating these neurobehavioural consequences of chronic sleep loss and to develop effective interventions to combat them.


Assuntos
Ansiedade/metabolismo , Hipocampo/metabolismo , Deficiências da Aprendizagem/metabolismo , Transtornos da Memória/metabolismo , Córtex Piriforme/metabolismo , Privação do Sono/metabolismo , Animais , Ansiedade/etiologia , Ansiedade/patologia , Doença Crônica , Hipocampo/patologia , Interleucina-1beta/metabolismo , Deficiências da Aprendizagem/etiologia , Deficiências da Aprendizagem/patologia , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Córtex Piriforme/patologia , Ratos , Ratos Wistar , Privação do Sono/complicações , Privação do Sono/patologia , Fator de Necrose Tumoral alfa/metabolismo
17.
Brain Res ; 1682: 36-43, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29307593

RESUMO

Sleep deprivation (SD) causes oxidative stress in the hippocampus and subsequent memory impairment. In this study, the effect of near-infrared (NIR) photobiomodulation (PBM) on learning and memory impairment induced by acute SD was investigated. The mice were subjected to an acute SD protocol for 72 h. Simultaneously, NIR PBM using a laser at 810 nm was delivered (once a day for 3 days) transcranially to the head to affect the entire brain of mice. The Barnes maze and the What-Where-Which task were used to assess spatial and episodic-like memories. The hippocampal levels of antioxidant enzymes and oxidative stress biomarkers were evaluated. The results showed that NIR PBM prevented cognitive impairment induced by SD. Moreover, NIR PBM therapy enhanced the antioxidant status and increased mitochondrial activity in the hippocampus of SD mice. Our findings revealed that hippocampus-related mitochondrial damage and extensive oxidative stress contribute to the occurrence of memory impairment. In contrast, NIR PBM reduced hippocampal oxidative damage, supporting the ability of 810 nm laser light to improve the antioxidant defense system and maintain mitochondrial survival. This confirms that non-invasive transcranial NIR PBM therapy ameliorates hippocampal dysfunction, which is reflected in enhanced memory function.


Assuntos
Hipocampo/metabolismo , Terapia com Luz de Baixa Intensidade/métodos , Transtornos da Memória , Estresse Oxidativo/fisiologia , Privação do Sono/complicações , Aldeídos/metabolismo , Animais , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Peroxidação de Lipídeos/fisiologia , Peroxidação de Lipídeos/efeitos da radiação , Masculino , Malondialdeído/metabolismo , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Transtornos da Memória/terapia , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/patologia , Estresse Oxidativo/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Privação do Sono/patologia , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Fatores de Tempo
18.
Life Sci ; 173: 94-106, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28214594

RESUMO

Exposure to Artificial Light At Night (ALAN) results in a disruption of the circadian system, which is deleterious to health. In industrialized countries, 75% of the total workforce is estimated to have been involved in shift work and night work. Epidemiologic studies, mainly of nurses, have revealed an association between sustained night work and a 50-100% higher incidence of breast cancer. The potential and multifactorial mechanisms of the effects include the suppression of melatonin secretion by ALAN, sleep deprivation, and circadian disruption. Shift and/or night work generally decreases the time spent sleeping, and it disrupts the circadian time structure. In the long run, this desynchronization is detrimental to health, as underscored by a large number of epidemiological studies that have uncovered elevated rates of several diseases, including cancer, diabetes, cardiovascular risks, obesity, mood disorders and age-related macular degeneration. It amounts to a public health issue in the light of the very substantial number of individuals involved. The IARC has classified shift work in group 2A of "probable carcinogens to humans" since "they involve a circadian disorganization". Countermeasures to the effects of ALAN, such as melatonin, bright light, or psychotropic drugs, have been proposed as a means to combat circadian clock disruption and improve adaptation to shift and night work. We review the evidence for the ALAN impacts on health. Furthermore, we highlight the importance of an in-depth mechanistic understanding to combat the detrimental properties of exposure to ALAN and develop strategies of prevention.


Assuntos
Ritmo Circadiano , Luz , Iluminação/efeitos adversos , Melatonina/metabolismo , Privação do Sono , Humanos , Privação do Sono/etiologia , Privação do Sono/metabolismo , Privação do Sono/patologia , Privação do Sono/fisiopatologia
19.
Behav Brain Res ; 322(Pt A): 9-17, 2017 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-28089853

RESUMO

Using a rat model of chronic sleep restriction (CSR) featuring periodic sleep deprivation with slowly rotating wheels (3h on/1h off), we previously observed that 99h of this protocol induced both homeostatic and allostatic (adaptive) changes in physiological and behavioural measures. Notably, the initial changes in sleep intensity and attention performance gradually adapted during CSR despite accumulating sleep loss. To identify brain regions involved in these responses, we used FosB/ΔFosB immunohistochemistry as a marker of chronic neuronal activation. Adult male rats were housed in motorized activity wheels and underwent the 3/1 CSR protocol for 99h, or 99h followed by 6 or 12days of recovery. Control rats were housed in home cages, locked activity wheels, or unlocked activity wheels that the animals could turn freely. Immunohistochemistry was conducted using an antibody that recognized both FosB and ΔFosB, and 24 brain regions involved in sleep/wake, autonomic, and limbic functions were examined. The number of darkly-stained FosB/ΔFosB-immunoreactive cells was increased immediately following 99h of CSR in 8/24 brain regions, including the medial preoptic and perifornical lateral hypothalamic areas, dorsomedial and paraventricular hypothalamic nuclei, and paraventricular thalamic nucleus. FosB/ΔFosB labeling was at control levels in all 8 brain areas following 6 or 12 recovery days, suggesting that most of the immunoreactivity immediately after CSR reflected FosB, the more transient marker of chronic neuronal activation. This region-specific induction of FosB/ΔFosB following CSR may be involved in the mechanisms underlying the allostatic changes in behavioural and physiological responses to CSR.


Assuntos
Encéfalo/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Privação do Sono/metabolismo , Animais , Encéfalo/patologia , Contagem de Células , Modelos Animais de Doenças , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos Wistar , Privação do Sono/patologia
20.
Mol Neurobiol ; 54(4): 3050-3061, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27037574

RESUMO

Modern lifestyle and sustained stress of professional commitments in the current societal set up often disrupts the normal sleep cycle and duration which is known to lead to cognitive impairments. In the present study, we report whether leaf extract of Withania somnifera (Ashwagandha) has potential neuroprotective role in acute stress of sleep deprivation. Experiments were performed on three groups of adult Wistar rats: group 1 (vehicle treated-undisturbed sleep [VUD]), group 2 (vehicle treated-sleep deprived [VSD]), and group 3 (ASH-WEX treated-sleep deprived [WSD]). Groups 1 and 2 received single oral feeding of vehicle and group 3 received ASH-WEX orally (140 mg/kg or 1 ml/250 g of body weight) for 15 consecutive days. Immediately after this regimen, animals from group 1 were allowed undisturbed sleep (between 6 a.m. and 6 p.m.), whereas rats of groups 2 and 3 were deprived of sleep during this period. We observed that WSD rats showed significant improvement in their performance in behavioral tests as compared to VSD group. At the molecular level, VSD rats showed acute change in the expression of proteins involved in synaptic plasticity, cell survival, and apoptosis in the hippocampus region of brain, which was suppressed by ASH-WEX treatment thus indicating decreased cellular stress and apoptosis in WSD group. This data suggest that Ashwagandha may be a potential agent to suppress the acute effects of sleep loss on learning and memory impairments and may emerge as a novel supplement to control SD-induced cognitive impairments.


Assuntos
Fármacos Neuroprotetores/uso terapêutico , Extratos Vegetais/uso terapêutico , Folhas de Planta/química , Privação do Sono/tratamento farmacológico , Withania/química , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Feminino , Homeostase/efeitos dos fármacos , Imuno-Histoquímica , Memória/efeitos dos fármacos , Modelos Biológicos , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Ratos Wistar , Privação do Sono/patologia , Privação do Sono/fisiopatologia , Estresse Fisiológico/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA