Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
BMC Psychiatry ; 24(1): 324, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664669

RESUMO

BACKGROUND: Methamphetamine (MA) abuse has resulted in a plethora of social issues. Sleep disturbance is a prominent issue about MA addiction, which serve as a risk factor for relapse, and the gut microbiota could play an important role in the pathophysiological mechanisms of sleep disturbances. Therefore, improving sleep quality can be beneficial for treating methamphetamine addiction, and interventions addressing the gut microbiota may represent a promising approach. METHOD: We recruited 70 MA users to investigate the associations between sleep quality and fecal microbiota by the Pittsburgh Sleep Quality Index (PSQI), which was divided into MA-GS (PSQI score < 7, MA users with good sleep quality, n = 49) and MA-BS group (PSQI score ≥ 7, MA users with bad sleep quality, n = 21). In addition, we compared the gut microbiota between the MA-GS and healthy control (HC, n = 38) groups. 16S rRNA sequencing was applied to identify the gut bacteria. RESULT: The study revealed that the relative abundances of the Thermoanaerobacterales at the order level differed between the MA-GS and MA-BS groups. Additionally, a positive correlation was found between the relative abundance of the genus Sutterella and daytime dysfunction. Furthermore, comparisons between MA users and HCs revealed differences in beta diversity and relative abundances of various bacterial taxa. CONCLUSION: In conclusion, the study investigated alterations in the gut microbiota among MA users. Furthermore, we demonstrated that the genus Sutterella changes may be associated with daytime dysfunction, suggesting that the genus Sutterella may be a biomarker for bad sleep quality in MA users.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas , Fezes , Microbioma Gastrointestinal , Metanfetamina , Qualidade do Sono , Humanos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Metanfetamina/efeitos adversos , Masculino , Adulto , Fezes/microbiologia , Feminino , RNA Ribossômico 16S/genética , Adulto Jovem , Transtornos do Sono-Vigília/microbiologia
2.
Brain Res Bull ; 180: 131-146, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35032622

RESUMO

Sleep is a universal physiological need in all species and is essential for the maintenance and recovery of various physiological functions of the body. In late years, the gut microbiota (GM), a vast and extraordinarily complex ecosystem located in human gastrointestinal tract that oversees an array of critical bodily functions, has become a popular focus among researchers. Accumulated evidences in this field have revealed that it exerts important roles in the regulation of some biological characteristics, especially metabolic, immunological and neurobehavioral functions. With the increasing comprehension of brain-gut axis, a bidirectional communication channel linking the brain and gut, the roles of GM in sleep are paid much attention to. Evidences have shown that the GM is essential for the maintenance of normal sleep physiology. In turn, it has also been demonstrated that the abnormal sleep patterns and duration affect the composition, diversity and function of the GM through the brain-gut-microbiota axis (BGMA). Present contributions have described several underlying factors that could be involved in the BGMA in sleep, such as the immune system, the vagus nerve, the neuroendocrine system, and bacterial metabolites. Furthermore, several interventions targeting the GM have been proved to be beneficial for amelioration of sleep problems. On this basis, in this review, we aim to explore the interaction between sleep and GM, and elucidate the therapeutic microbiota manipulations with potential promoting effects on sleep quality.


Assuntos
Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia , Transtornos do Sono-Vigília , Sono/fisiologia , Humanos , Transtornos do Sono-Vigília/imunologia , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/terapia
3.
PLoS One ; 16(9): e0255323, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34495955

RESUMO

Sleep disorders are increasingly being characterized in modern society as contributing to a host of serious medical problems, including obesity and metabolic syndrome. Changes to the microbial community in the human gut have been reportedly associated with many of these cardiometabolic outcomes. In this study, we investigated the impact of sleep length on the gut microbiota in a large cohort of 655 participants of African descent, aged 25-45, from Ghana, South Africa (SA), Jamaica, and the United States (US). The sleep duration was self-reported via a questionnaire. Participants were classified into 3 sleep groups: short (<7hrs), normal (7-<9hrs), and long (≥9hrs). Forty-seven percent of US participants were classified as short sleepers and 88% of SA participants as long sleepers. Gut microbial composition analysis (16S rRNA gene sequencing) revealed that bacterial alpha diversity negatively correlated with sleep length (p<0.05). Furthermore, sleep length significantly contributed to the inter-individual beta diversity dissimilarity in gut microbial composition (p<0.01). Participants with both short and long-sleep durations exhibited significantly higher abundances of several taxonomic features, compared to normal sleep duration participants. The predicted relative proportion of two genes involved in the butyrate synthesis via lysine pathway were enriched in short sleep duration participants. Finally, co-occurrence relationships revealed by network analysis showed unique interactions among the short, normal and long duration sleepers. These results suggest that sleep length in humans may alter gut microbiota by driving population shifts of the whole microbiota and also specific changes in Exact Sequence Variants abundance, which may have implications for chronic inflammation associated diseases. The current findings suggest a possible relationship between disrupted sleep patterns and the composition of the gut microbiota. Prospective investigations in larger and more prolonged sleep researches and causally experimental studies are needed to confirm these findings, investigate the underlying mechanism and determine whether improving microbial homeostasis may buffer against sleep-related health decline in humans.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal/fisiologia , Transtornos do Sono-Vigília/microbiologia , Sono/fisiologia , Adulto , Bactérias/genética , Bactérias/isolamento & purificação , Estudos de Coortes , Fezes/microbiologia , Feminino , Gana , Humanos , Jamaica , Masculino , Pessoa de Meia-Idade , RNA Ribossômico 16S/genética , Análise de Sequência de DNA/métodos , África do Sul , Inquéritos e Questionários , Estados Unidos
4.
Sci Rep ; 11(1): 16313, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381098

RESUMO

The first night effect (FNE) is a type of sleep disturbance caused by an unfamiliar environment, which leads to difficulty falling asleep and reduced sleep duration. Previously, we reported that Lactobacillus fermentum PS150 (PS150) improves sleep conditions in a pentobarbital-induced sleep mouse model. In this study, we aimed to evaluate the effect of PS150 on the FNE in mice. Briefly, mice were implanted with electrodes and orally administered PS150 for four weeks, and then the FNE was induced by cage changing. Analysis of polysomnographic signals revealed that intervention with PS150 restored non-rapid eye movement (NREM) sleep length under the FNE. Compared to diphenhydramine, a commonly used sleep aid, PS150 had no unwanted side effects, such as rapid eye movement (REM) sleep deprivation and fragmented sleep. Moreover, temporal analysis revealed that PS150 efficiently reduced both sleep latency and time spent restoring normal levels of REM sleep. Taken together, these results suggest that PS150 efficiently ameliorates sleep disturbance caused by the FNE. Additionally, V3-V4 16S rRNA sequencing revealed significant increases in Erysipelotrichia, Actinobacteria, and Coriobacteriia in fecal specimens of the PS150-treated group, indicating that PS150 induces gut microbiota remodeling.


Assuntos
Limosilactobacillus fermentum/fisiologia , Sono REM/fisiologia , Animais , Modelos Animais de Doenças , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pentobarbital/farmacologia , Polissonografia/métodos , RNA Ribossômico 16S/genética , Privação do Sono/induzido quimicamente , Privação do Sono/microbiologia , Privação do Sono/fisiopatologia , Distúrbios do Início e da Manutenção do Sono/induzido quimicamente , Distúrbios do Início e da Manutenção do Sono/microbiologia , Distúrbios do Início e da Manutenção do Sono/fisiopatologia , Transtornos do Sono-Vigília/induzido quimicamente , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/fisiopatologia , Sono REM/efeitos dos fármacos
5.
Life Sci ; 283: 119851, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34324916

RESUMO

Sleep disorders frequently comorbid with several cardiovascular diseases (CVDs), attracting increasing scientific attention and interest. Sleep disorders include insomnia, sleep-disordered breathing, restless legs syndrome, etc. It is well known that inflammation, sympathetic activation, and endothelial dysfunction play critical roles in sleep disorders, all of which are predisposing factors for CVDs. The comorbidity of sleep disorders and CVDs may have a bidirectional relationship. Patients with CVDs may have a high incidence of sleep disorders and vice versa. This review focused on the comorbidity of sleep disorders and CVDs and discussed the potential pathophysiological mechanisms and therapeutic strategies. In addition to the existing mechanisms, this review summarized novel potential mechanisms underlying comorbidities, such as gut microbiota, orexin, and extracellular vesicles, which may provide a theoretical basis for further basic research and clinical investigations on improving therapeutic outcomes.


Assuntos
Doenças Cardiovasculares , Microbioma Gastrointestinal , Transtornos do Sono-Vigília , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/microbiologia , Doenças Cardiovasculares/terapia , Vesículas Extracelulares/metabolismo , Humanos , Orexinas/metabolismo , Transtornos do Sono-Vigília/complicações , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/terapia
6.
Biol Res Nurs ; 23(1): 31-41, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32700552

RESUMO

PURPOSE: To examine a) whether there are significant differences in the severity of symptoms of fatigue, sleep disturbance, or depression between patients with rectal cancer who develop co-occurring symptoms and those with no symptoms before and at the end of chemotherapy and radiation therapy (CRT); b) differences in gut microbial diversity between those with co-occurring symptoms and those with no symptoms; and c) whether before-treatment diversity measurements and taxa abundances can predict co-occurrence of symptoms. METHODS: Stool samples and symptom ratings were collected from 31 patients with rectal cancer prior to and at the end of (24-28 treatments) CRT. Descriptive statistics were computed and the Mann-Whitney U test was performed for symptoms. Gut microbiome data were analyzed using R's vegan package software. RESULTS: Participants with co-occurring symptoms reported greater severity of fatigue at the end of CRT than those with no symptoms. Bacteroides and Blautia2 abundances differed between participants with co-occurring symptoms and those with no symptoms. Our random forest classification (unsupervised learning algorithm) predicted participants who developed co-occurring symptoms with 74% accuracy, using specific phylum, family, and genera abundances as predictors. CONCLUSION: Our preliminary results point to an association between the gut microbiota and co-occurring symptoms in rectal cancer patients and serves as a first step in potential identification of a microbiota-based classifier.


Assuntos
Microbioma Gastrointestinal , Neoplasias Retais/microbiologia , Neoplasias Retais/terapia , Idoso , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Quimiorradioterapia , Depressão/microbiologia , Fadiga/microbiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudo de Prova de Conceito , Transtornos do Sono-Vigília/microbiologia
7.
Nutrients ; 11(8)2019 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-31405122

RESUMO

Short-term administration of Lactobacillus gasseri CP2305 improves stress-associated symptoms and clinical symptoms in healthy young adults and in patients with irritable bowel syndrome, respectively. We evaluated the efficacy and health benefits of the long-term use of a tablet containing heat-inactivated, washed Lactobacillus gasseri CP2305 (CP2305) in healthy young adults. Sixty Japanese medical students (41 men and 19 women) preparing for the national examination for medical practitioners ingested CP2305-containing or placebo tablets once daily for 24 weeks. Intake of the CP2305 tablet significantly reduced anxiety and sleep disturbance relative to placebo, as quantitated by the Spielberger State-Trait Anxiety Inventory and the Pittsburgh Sleep Quality Index. Single-channel sleep electroencephalograms show that CP2305 significantly shortened sleep latency and wake time after sleep onset and increased the delta power ratio in the first sleep cycle. CP2305 also significantly lowered salivary chromogranin A levels compared with placebo. Furthermore, 16S rRNA gene sequencing of participant feces demonstrated that CP2305 administration attenuated the stress-induced decline of Bifidobacterium spp. and the stress-induced elevation of Streptococcus spp. We conclude that the long-term use of CP2305-containing tablets may improve the mental state, sleep quality, and gut microbiota of healthy adults under stressful conditions.


Assuntos
Lactobacillus gasseri , Probióticos/administração & dosagem , Estresse Psicológico/microbiologia , Estresse Psicológico/terapia , Adulto , Bifidobacterium/metabolismo , Cromogranina A/metabolismo , Doença Crônica , Método Duplo-Cego , Feminino , Microbioma Gastrointestinal , Voluntários Saudáveis , Humanos , Masculino , RNA Ribossômico 16S/análise , Saliva/química , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/psicologia , Transtornos do Sono-Vigília/terapia , Streptococcus/metabolismo , Estresse Psicológico/psicologia , Estudantes de Medicina/psicologia , Comprimidos , Resultado do Tratamento , Adulto Jovem
9.
Psychosom Med ; 81(8): 704-710, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30308579

RESUMO

Within a couple, partners influence each other's mental and physical health. This review focuses on how couples' relationships, the partners' individual and joint vulnerabilities, and their health behaviors influence health through changes in the gut microbiota, metabolism, and immune function. Couples' shared stressors and emotions and their intertwined lifestyles and routines serve to promote common disease risks in part through parallel changes in their gut microbiotas. Marital discord, stress, and depression have strong bidirectional links, fueling one another. Chronic marital stress and depression can elevate the risk for obesity, metabolic syndrome, and cardiovascular disease by altering resting energy expenditure, insulin production, and triglyceride responses after unhealthy meals. During stressful times, health behaviors typically suffer-and sleep disturbances, poor diets, and sedentary behavior all influence these metabolic pathways while also promoting gut dysbiosis. Dysbiosis increases intestinal permeability (gut leakiness), providing a mechanistic pathway from marital distress and depression to heightened inflammation and accelerated aging. Age-related changes in the gut microbiota's composition and gut leakiness foster immunosenescence, as well as the progression of inflamm-aging; these age-related risks may be altered by stress and depression, diet, sleep, exercise habits, and developmental shifts in emotion regulation strategies. Consideration of the strong mutual influences that partners have on each other's mood and health behaviors, as well as the biological pathways that underlie these influences, provides a new way to view marriage's health implications.


Assuntos
Senilidade Prematura/etiologia , Envelhecimento/psicologia , Disbiose/etiologia , Emoções , Microbioma Gastrointestinal , Comportamentos Relacionados com a Saúde , Casamento , Senilidade Prematura/microbiologia , Senilidade Prematura/fisiopatologia , Senilidade Prematura/psicologia , Ritmo Circadiano , Depressão/etiologia , Depressão/microbiologia , Depressão/fisiopatologia , Depressão/psicologia , Dieta , Suscetibilidade a Doenças , Disbiose/fisiopatologia , Disbiose/psicologia , Emoções/fisiologia , Exercício Físico , Conflito Familiar/psicologia , Feminino , Microbioma Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Hábitos , Comportamentos Relacionados com a Saúde/fisiologia , Humanos , Imunidade , Inflamação/etiologia , Inflamação/microbiologia , Inflamação/fisiopatologia , Inflamação/psicologia , Estilo de Vida , Masculino , Casamento/psicologia , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/fisiopatologia , Transtornos do Sono-Vigília/psicologia , Estresse Psicológico/etiologia , Estresse Psicológico/microbiologia , Estresse Psicológico/fisiopatologia , Estresse Psicológico/psicologia
10.
Sci Rep ; 8(1): 16770, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30425273

RESUMO

Tonsil and adenoid-tissue hypertrophy (AH) is the most common cause of pediatric sleep-disordered breathing (SDB), with AH possibly initiated by repeated exposure to infectious agents or allergens. Here, we evaluated IL-17A activity in adenoid tissue from children with SDB and its association with AH and pneumococcal carriage. Thirty-five children (aged 3-12 years) with SDB and receiving adenoidectomy and tonsillectomy were enrolled. During surgery, nasopharyngeal carriage was determined by bacterial culture and multiplex PCR via nasopharyngeal swab, and adenoid samples were collected. IL-17A and associated cytokine expression was evaluated by real-time PCR and western blotting. The mRNA analysis showed that IL-17A level, IL-17A:IL-10 ratio, and RAR-related orphan receptor-γt:forkhead box P3 ratio were significantly higher in adenoid tissues with AH, as were IL-17A level and IL-17A:IL-10 ratio in adenoid tissues with pneumococcal carriage. Additionally, pneumococcal carriage was more common in nasopharyngeal adenoids from patients without AH than those with AH. IL-17A was upregulated in adenoid tissues from patients with AH and with pneumococcal carriage. These results suggested that pneumococcal carriage initiates an IL-17A-mediated immune response in nasopharyngeal adenoids, which might be associated with AH in patients with SDB.


Assuntos
Tonsila Faríngea/metabolismo , Regulação da Expressão Gênica , Interleucina-17/metabolismo , Respiração , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/fisiopatologia , Streptococcus pneumoniae/fisiologia , Tonsila Faríngea/microbiologia , Tonsila Faríngea/patologia , Criança , Pré-Escolar , Feminino , Humanos , Hipertrofia , Interleucina-17/genética , Masculino , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/patologia
11.
PLoS One ; 13(7): e0198288, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30020932

RESUMO

The microbiome plays a vital role in human health and disease. Interaction between human hosts and the microbiome occurs through a number of mechanisms, including transcriptomic regulation by microRNA (miRNA). In animal models, circadian variations in miRNA and microbiome elements have been described, but patterns of co-expression and potential diurnal interaction in humans have not. We investigated daily oscillations in salivary miRNA and microbial RNA to explore relationships between these components of the gut-brain-axis and their implications in human health. Nine subjects provided 120 saliva samples at designated times, on repeated days. Samples were divided into three sets for exploration and cross-validation. Identification and quantification of host miRNA and microbial RNA was performed using next generation sequencing. Three stages of statistical analyses were used to identify circadian oscillators: 1) a two-way analysis of variance in the first two sample sets identified host miRNAs and microbial RNAs whose abundance varied with collection time (but not day); 2) multivariate modeling identified subsets of these miRNAs and microbial RNAs strongly-associated with collection time, and evaluated their predictive ability in an independent hold-out sample set; 3) regulation of circadian miRNAs and microbial RNAs was explored in data from autistic children with disordered sleep (n = 77), relative to autistic peers with typical sleep (n = 63). Eleven miRNAs and 11 microbial RNAs demonstrated consistent diurnal oscillation across sample sets and accurately predicted collection time in the hold-out set. Associations among five circadian miRNAs and four circadian microbial RNAs were observed. We termed the 11 miRNAs CircaMiRs. These CircaMiRs had 1,127 predicted gene targets, with enrichment for both circadian gene targets and metabolic signaling processes. Four CircaMiRs had "altered" expression patterns among children with disordered sleep. Thus, novel and correlated circadian oscillations in human miRNA and microbial RNA exist and may have distinct implications in human health and disease.


Assuntos
Transtorno Autístico/genética , MicroRNAs/genética , Saliva/química , Transtornos do Sono-Vigília/genética , Adolescente , Adulto , Transtorno Autístico/microbiologia , Transtorno Autístico/fisiopatologia , Criança , Pré-Escolar , Relógios Circadianos/genética , Feminino , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno/genética , Humanos , Masculino , MicroRNAs/química , MicroRNAs/isolamento & purificação , Microbiota/genética , Pessoa de Meia-Idade , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , RNA Bacteriano/química , RNA Bacteriano/genética , RNA Bacteriano/isolamento & purificação , Saliva/microbiologia , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/fisiopatologia , Transcriptoma/genética , Adulto Jovem
13.
Int Rev Neurobiol ; 131: 207-225, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27793219

RESUMO

Sleep is profoundly altered during the course of infectious diseases. The typical response to infection includes an initial increase in nonrapid eye movement sleep (NREMS) followed by an inhibition in NREMS. REMS is inhibited during infections. Bacterial cell wall components, such as peptidoglycan and lipopolysaccharide, macrophage digests of these components, such as muramyl peptides, and viral products, such as viral double-stranded RNA, trigger sleep responses. They do so via pathogen-associated molecular pattern recognition receptors that, in turn, enhance cytokine production. Altered sleep and associated sleep-facilitated fever responses are likely adaptive responses to infection. Normal sleep in physiological conditions may also be influenced by gut microbes because the microbiota is affected by circadian rhythms, stressors, diet, and exercise. Furthermore, sleep loss enhances translocation of viable bacteria from the intestine, which provides another means by which sleep-microbe interactions impact neurobiology.


Assuntos
Microbioma Gastrointestinal/fisiologia , Transtornos do Sono-Vigília , Sono/fisiologia , Animais , Citocinas/metabolismo , Humanos , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/patologia
14.
Environ Res ; 137: 357-63, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25601739

RESUMO

BACKGROUND: Exposure to mould or dampness at home has been associated with adverse respiratory effects in all age groups. This exposure has also been related to insomnia in adults. We aimed to investigate the association between exposure to visible mould or dampness at home and sleep problems in children. METHODS: The study population consisted of 1719 10-year-old children from the German population-based birth cohort LISAplus with available data on current mould or dampness at home and sleep problems. The presence of visible mould or dampness at home was assessed by questionnaire. Parent-reported sleep problems of their child were analysed by four binary variables: presence of any sleep problems, problems to fall asleep, problems sleeping through the night and a 24h sleep time of less than 9h. Logistic regression models adjusted for study centre, sex, age and level of parental education were applied to examine the association between visible mould or dampness at home and sleep problems. Sensitivity analyses included a further adjustment for bedroom sharing and subgroup analyses in children without current allergic diseases. RESULTS: Thirteen percent of parents reported visible mould or dampness at home. We observed increased risks for all four sleep problem variables for children exposed to visible mould or dampness at home. Results were significant for any sleep problems (odds ratio (OR)=1.77 (95%-confidence interval (CI): 1.21-2.60), problems sleeping through the night (OR=2.52(1.27-5.00) and a short sleep time (OR=1.68(1.09-2.61)). While a further adjustment for bedroom sharing and the exclusion of children with asthma or eczema led to similar results, only the association with a short sleep time was still present in children without allergic rhinoconjunctivitis. CONCLUSION: Our data suggests that visible mould or dampness at home might negatively influence sleep in children. The influence of allergic rhinoconjunctivitis on this association needs to be investigated in future studies.


Assuntos
Asma/epidemiologia , Eczema/epidemiologia , Exposição Ambiental , Fungos/fisiologia , Umidade/efeitos adversos , Rinite Alérgica/epidemiologia , Transtornos do Sono-Vigília/epidemiologia , Asma/complicações , Asma/microbiologia , Criança , Eczema/complicações , Eczema/microbiologia , Feminino , Alemanha/epidemiologia , Habitação , Humanos , Hipersensibilidade/epidemiologia , Masculino , Rinite Alérgica/complicações , Rinite Alérgica/microbiologia , Transtornos do Sono-Vigília/microbiologia
15.
Am J Physiol Regul Integr Comp Physiol ; 301(5): R1467-78, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21900639

RESUMO

Sepsis is a systemic immune response to infection that may result in multiple organ failure and death. Polymicrobial infections remain a serious clinical problem, and in the hospital, sepsis is the number-one noncardiac killer. Although the central nervous system may be one of the first systems affected, relatively little effort has been made to determine the impact of sepsis on the brain. In this study, we used the cecal ligation and puncture (CLP) model to determine the extent to which sepsis alters sleep, the EEG, and brain temperature (Tbr) of rats. Sepsis increases the amount of time rats spend in non-rapid eye movement sleep (NREMS) during the dark period, but not during the light period. Rapid eye movements sleep (REMS) of septic rats is suppressed for about 24 h following CLP surgery, after which REMS increases during dark periods for at least three nights. The EEG is dramatically altered shortly after sepsis induction, as evidenced by reductions in slow-frequency components. Furthermore, sleep is fragmented, indicating that the quality of sleep is diminished. Effects on sleep, the EEG, and Tbr persist for at least 84 h after sepsis induction, the duration of our recording period. Immunohistochemical assays focused on brain stem mechanisms responsible for alterations in REMS, as little information is available concerning infection-induced suppression of this sleep stage. Our immunohistochemical data suggest that REMS suppression after sepsis onset may be mediated, in part, by the brain stem GABAergic system. This study demonstrates for the first time that sleep and EEG patterns are altered during CLP-induced sepsis. These data suggest that the EEG may serve as a biomarker for sepsis onset. These data also contribute to our knowledge of potential mechanisms, whereby infections alter sleep and other central nervous system functions.


Assuntos
Encéfalo/fisiopatologia , Sepse/complicações , Fases do Sono , Transtornos do Sono-Vigília/etiologia , Animais , Comportamento Animal , Temperatura Corporal , Encéfalo/metabolismo , Encéfalo/microbiologia , Ceco/microbiologia , Ceco/cirurgia , Ritmo Circadiano , Modelos Animais de Doenças , Eletroencefalografia , Glutamato Descarboxilase/metabolismo , Imuno-Histoquímica , Ligadura , Masculino , Fotoperíodo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Punções , Ratos , Ratos Sprague-Dawley , Sepse/metabolismo , Sepse/microbiologia , Sepse/fisiopatologia , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/microbiologia , Transtornos do Sono-Vigília/fisiopatologia , Sono REM , Fatores de Tempo , Ácido gama-Aminobutírico/metabolismo
16.
Przegl Epidemiol ; 58(3): 445-50, 2004.
Artigo em Polonês | MEDLINE | ID: mdl-15730008

RESUMO

The aim of the study was to evaluate relationship between initial manifestation of Lyme borreliosis and further course of the disease. Files and questionnaires regarding present and past course of the disease were evaluated in 138 residents of Bialowieza village, suffering previously from Lyme borreliosis. Present arthralgia was the most frequently reported by persons who initially were diagnosed as suffering from Lyme arthritis. Ischialgia and brachialgia were the most prevalent in patients with previous neuroboreliosis. Psychiatric evaluation demonstrated significantly more frequent prevalence of sleep and cognitive disturbances in persons with previously diagnosed neuroboreliosis. Persons with initial joint manifestations of Lyme borreliosis more frequently report arthralgia in further follow-up. Primary manifestation of the disease as neuroboreliosis increase risk of subsequent disturbances of sleep and cognition.


Assuntos
Grupo Borrelia Burgdorferi/isolamento & purificação , Nível de Saúde , Doença de Lyme/complicações , Doença de Lyme/diagnóstico , Adulto , Animais , Artralgia/microbiologia , Artrite Infecciosa/microbiologia , Neurite do Plexo Braquial/microbiologia , Transtornos Cognitivos/microbiologia , Feminino , Seguimentos , Humanos , Mordeduras e Picadas de Insetos/complicações , Doença de Lyme/epidemiologia , Masculino , Pessoa de Meia-Idade , Polônia/epidemiologia , Prevalência , Fatores de Risco , Ciática/microbiologia , Transtornos do Sono-Vigília/microbiologia , Inquéritos e Questionários , Carrapatos , Fatores de Tempo
17.
Brain Res ; 832(1-2): 54-62, 1999 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-10375652

RESUMO

Microglial cells play a key role in the events triggered by infection, injury or degeneration in the central nervous system not only as scavenger cells but also as immune effector elements. We analyzed the features and distribution of cells of the microglia/macrophage lineage with OX-42 and ED-1 immunohistochemistry in the brain of experimental rats infected with the extracellular parasite Trypanosoma brucei. Such experimental infection provides a rat model of sleeping sickness or African trypanosomiasis, and is hallmarked in its advanced stages by severe alterations of the animals' sleep structure. In infected rats a remarkable activation of microglia, revealed by OX-42 immunoreactivity, became evident in the 3rd week post-infection in periventricular and subpial brain regions, with a prevalence in the hypothalamus. These features were concomitant with the onset of sleep anomalies, monitored with electroencephalographic recordings. Microglia activation increased in the following weeks, paralleling the progressive alterations of sleep parameters, and was most marked in the terminal stages of the infection, corresponding to the 6th-7th weeks. In addition, ED-1-immunoreactive macrophages and ramified microglia, confined to hypothalamic periventricular and basal regions, were evident after 4 weeks of disease. Degeneration of neuronal perikarya was not detected histologically in the infected brains at any time point. These data provide evidence for a reaction of microglia and macrophages in the brain of trypanosome-infected rats, and point out a selective distribution of these activated cells. The findings are discussed in relation to the animals' sleep disorder during trypanosome infection.


Assuntos
Ativação de Macrófagos , Microglia/química , Transtornos do Sono-Vigília/metabolismo , Trypanosoma brucei brucei , Tripanossomíase Africana/metabolismo , Animais , Linhagem da Célula , Modelos Animais de Doenças , Eletroencefalografia , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley , Transtornos do Sono-Vigília/microbiologia , Tripanossomíase Africana/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA