Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Diabetologia ; 67(6): 1114-1121, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38413436

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to explore whether diabetic retinopathy is associated with alterations of the circadian system, and to examine the role of reduced intrinsically photosensitive retinal ganglion cell (ipRGC) function. METHODS: Participants with type 2 diabetes, with diabetic retinopathy (n=14) and without diabetic retinopathy (n=9) underwent 24 h blood sampling for melatonin and cortisol under controlled laboratory conditions. ipRGC function was inferred from the post-illumination pupil response (PIPR). Habitual sleep duration, efficiency and variability were assessed by actigraphy. RESULTS: Participants with diabetic retinopathy compared to participants without diabetic retinopathy had smaller PIPR (p=0.007), lower 24 h serum melatonin output (p=0.042) and greater day-to-day sleep variability (p=0.012). By contrast, 24 h cortisol profiles, sleep duration and efficiency were similar in both groups. Six individuals with diabetic retinopathy had no detectable dim-light melatonin onset. PIPR correlated with 24 h mean melatonin levels (r=0.555, p=0.007). CONCLUSIONS/INTERPRETATION: ipRCG dysfunction in diabetic retinopathy is associated with disruptions of the 24 h melatonin rhythm, suggesting circadian dysregulation in diabetic retinopathy.


Subject(s)
Circadian Rhythm , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Melatonin , Retinal Ganglion Cells , Humans , Melatonin/blood , Melatonin/metabolism , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/blood , Diabetic Retinopathy/physiopathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Male , Female , Cross-Sectional Studies , Middle Aged , Circadian Rhythm/physiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Aged , Hydrocortisone/blood , Hydrocortisone/metabolism , Sleep/physiology , Adult
2.
Sleep Med Rev ; 74: 101891, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38118339

ABSTRACT

Diabetic retinopathy (DR) is one of the most prevalent microvascular diabetic complications. Poor sleep health and obstructive sleep apnea (OSA) are risk factors for diabetes and poor glycemic control. Recent studies have suggested associations between poor sleep health/OSA and DR. Furthermore, there have been suggestions of melatonin dysregulation in the context of DR. We conducted a systematic review and meta-analysis exploring the associations between multidimensional sleep health (duration, satisfaction, efficiency, timing/regularity and alertness), OSA and melatonin with DR. Forty-two studies were included. Long, but not short sleep, was significantly associated with DR, OR 1.41 (95%CI 1.21, 1.64). Poor sleep satisfaction was also significantly associated with DR, OR 2.04 (1.41, 2.94). Sleep efficiency and alertness were not associated with DR, while the evidence on timing/regularity was scant. Having OSA was significantly associated with having DR, OR 1.34 (1.07, 1.69). Further, those with DR had significantly lower melatonin/melatonin metabolite levels than those without DR, standardized mean difference -0.94 (-1.44, -0.44). We explored whether treating OSA with continuous positive airway pressure (CPAP) led to improvement in DR (five studies). The results were mixed among studies, but potential benefits were observed in some. This review highlights the association between poor multidimensional sleep health and DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Melatonin , Sleep Apnea, Obstructive , Sleep Initiation and Maintenance Disorders , Humans , Sleep , Risk Factors , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/therapy , Sleep Initiation and Maintenance Disorders/complications , Continuous Positive Airway Pressure
3.
J Sleep Res ; : e13989, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37414725

ABSTRACT

Sleep irregularity and variability have been shown to be detrimental to cardiometabolic health. The present pilot study explored if higher day-to-day sleep irregularity and variability were associated with systemic inflammation, as assessed by high-sensitivity C-reactive protein, in type 2 diabetes. Thirty-five patients with type 2 diabetes (mean age 54.3 years, 54.3% female) who were not shift-workers participated. The presence of diabetic retinopathy was determined. The standard deviation of sleep duration and sleep midpoint across all recorded nights were used to quantify sleep variability and regularity, respectively, assessed by 14-day actigraphy. The presence and severity of sleep apnea were assessed using an overnight home monitor. Low-density lipoprotein, haemoglobin A1C and high-sensitivity C-reactive protein were collected. Multiple regression analysis using natural-log-transformed values was performed to establish an independent association between sleep variability and high-sensitivity C-reactive protein. Twenty-two (62.9%) patients had diabetic retinopathy. The median (interquartile range) of high-sensitivity C-reactive protein was 2.4 (1.4, 4.6) mg L-1 . Higher sleep variability was significantly associated with higher high-sensitivity C-reactive protein (r = 0.342, p = 0.044), as was haemoglobin A1C (r = 0.431, p = 0.010) and low-density lipoprotein (r = 0.379, p = 0.025), but not sleep regularity, sleep apnea severity or diabetic retinopathy. Multiple regression analysis showed that higher sleep variability (B = 0.907, p = 0.038) and higher HbA1c (B = 1.519, p = 0.035), but not low-density lipoprotein, contributed to higher high-sensitivity C-reactive protein. In conclusion, higher sleep variability in patients with type 2 diabetes who were not shift-workers was independently associated with higher systemic inflammation, conferring increased cardiovascular risk. Whether sleep interventions to reduce sleep variability can reduce systemic inflammation and improve cardiometabolic health should be investigated.

4.
Am J Clin Nutr ; 116(6): 1877-1900, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36055772

ABSTRACT

Precision nutrition is an emerging concept that aims to develop nutrition recommendations tailored to different people's circumstances and biological characteristics. Responses to dietary change and the resulting health outcomes from consuming different diets may vary significantly between people based on interactions between their genetic backgrounds, physiology, microbiome, underlying health status, behaviors, social influences, and environmental exposures. On 11-12 January 2021, the National Institutes of Health convened a workshop entitled "Precision Nutrition: Research Gaps and Opportunities" to bring together experts to discuss the issues involved in better understanding and addressing precision nutrition. The workshop proceeded in 3 parts: part I covered many aspects of genetics and physiology that mediate the links between nutrient intake and health conditions such as cardiovascular disease, Alzheimer disease, and cancer; part II reviewed potential contributors to interindividual variability in dietary exposures and responses such as baseline nutritional status, circadian rhythm/sleep, environmental exposures, sensory properties of food, stress, inflammation, and the social determinants of health; part III presented the need for systems approaches, with new methods and technologies that can facilitate the study and implementation of precision nutrition, and workforce development needed to create a new generation of researchers. The workshop concluded that much research will be needed before more precise nutrition recommendations can be achieved. This includes better understanding and accounting for variables such as age, sex, ethnicity, medical history, genetics, and social and environmental factors. The advent of new methods and technologies and the availability of considerably more data bring tremendous opportunity. However, the field must proceed with appropriate levels of caution and make sure the factors listed above are all considered, and systems approaches and methods are incorporated. It will be important to develop and train an expanded workforce with the goal of reducing health disparities and improving precision nutritional advice for all Americans.


Subject(s)
Evidence Gaps , Nutritional Status , Humans , United States , Precision Medicine/methods , Diet , National Institutes of Health (U.S.) , Nutrigenomics
5.
Thyroid ; 32(2): 196-205, 2022 02.
Article in English | MEDLINE | ID: mdl-34641706

ABSTRACT

Background: L-triiodothyronine (LT3) has been increasingly used in combination with levothyroxine in the treatment of hypothyroidism. A metal coordinated form of LT3, known as poly-zinc-liothyronine (PZL), avoided in rats the typical triiodothyronine (T3) peak seen after oral administration of LT3. Objectives: To evaluate in healthy volunteers (i) the pharmacokinetics (PK) of PZL-derived T3 after a single dose, (ii) the pharmacodynamics of PZL-derived T3, (iii) incidence of adverse events, and (iv) exploratory analysis of the sleep patterns after LT3, PZL, or placebo (PB) administration. Methods: Twelve healthy volunteers 18-50 years of age were recruited for a Phase 1, double-blind, randomized, single-dose PB-controlled, crossover study to compare PZL against LT3 or PB. Subjects were admitted three separate times to receive a randomly assigned capsule containing PB, 50 µg LT3, or 50 µg PZL, and were observed for 48 hours. A 2-week washout period separated each admission. Results: LT3-derived serum T3 levels exhibited the expected profile, with a Tmax at 2 hours and return to basal levels by 24-36 hours. PZL-derived serum T3 levels exhibited ∼30% lower Cmax that was 1 hour delayed and extended into a plateau that lasted up to 6 hours. This was followed by a lower but much longer plateau; by 24 hours serum T3 levels still exceeded ½ of Cmax. Thyrotropin levels were similarly reduced in both groups. Conclusion: PZL possesses the necessary properties to achieve a much improved T3 PK. PZL is on track to provide hypothyroid patients with stable levels of serum T3.


Subject(s)
Triiodothyronine/administration & dosage , Triiodothyronine/pharmacokinetics , Zinc/administration & dosage , Adolescent , Adult , Double-Blind Method , Drug Therapy, Combination , Female , Hormone Replacement Therapy , Humans , Hypothyroidism/drug therapy , Male , Middle Aged , Young Adult
6.
Ann N Y Acad Sci ; 1506(1): 18-34, 2021 12.
Article in English | MEDLINE | ID: mdl-34341993

ABSTRACT

The human circadian system consists of the master clock in the suprachiasmatic nuclei of the hypothalamus as well as in peripheral molecular clocks located in organs throughout the body. This system plays a major role in the temporal organization of biological and physiological processes, such as body temperature, blood pressure, hormone secretion, gene expression, and immune functions, which all manifest consistent diurnal patterns. Many facets of modern life, such as work schedules, travel, and social activities, can lead to sleep/wake and eating schedules that are misaligned relative to the biological clock. This misalignment can disrupt and impair physiological and psychological parameters that may ultimately put people at higher risk for chronic diseases like cancer, cardiovascular disease, and other metabolic disorders. Understanding the mechanisms that regulate sleep circadian rhythms may ultimately lead to insights on behavioral interventions that can lower the risk of these diseases. On February 25, 2021, experts in sleep, circadian rhythms, and chronobiology met virtually for the Keystone eSymposium "Sleep & Circadian Rhythms: Pillars of Health" to discuss the latest research for understanding the bidirectional relationships between sleep, circadian rhythms, and health and disease.


Subject(s)
Circadian Rhythm/physiology , Congresses as Topic/trends , Meals/physiology , Research Report , Sleep/physiology , Animals , Blood Pressure/physiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/psychology , Circadian Clocks/physiology , Humans , Meals/psychology , Neoplasms/genetics , Neoplasms/physiopathology , Neoplasms/psychology , Risk Factors
7.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161260

ABSTRACT

Individuals who are minoritized as a result of race, sexual identity, gender, or socioeconomic status experience a higher prevalence of many diseases. Understanding the biological processes that cause and maintain these socially driven health inequities is essential for addressing them. The gut microbiome is strongly shaped by host environments and affects host metabolic, immune, and neuroendocrine functions, making it an important pathway by which differences in experiences caused by social, political, and economic forces could contribute to health inequities. Nevertheless, few studies have directly integrated the gut microbiome into investigations of health inequities. Here, we argue that accounting for host-gut microbe interactions will improve understanding and management of health inequities, and that health policy must begin to consider the microbiome as an important pathway linking environments to population health.


Subject(s)
Gastrointestinal Microbiome , Health Status Disparities , Disease , Health , Humans , Mental Health , Publications
8.
Curr Opin Endocr Metab Res ; 17: 15-19, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33283071

ABSTRACT

In this literature review, we discuss the importance of adequate sleep and the various effects of suboptimal sleep on weight maintenance and metabolic health specifically for adolescents. Two major contributors to adolescents experiencing decreased sleep duration and quality, and thus increasing the risk for developing metabolic syndrome in adolescence as well as later in adulthood, are increased electronic screen time particularly at night and early school start times. The less time adolescents spend sleeping, the less quality sleep they obtain, and the greater the disruption of endocrine hormone function. As another consequence, adolescents are more prone to making poor food choices, from choosing relatively nutrient-poor foods to consuming excess calories without necessarily increasing their energy expenditure. These choices put adolescents at greater risk for becoming obese throughout their lifespan.

9.
Obes Sci Pract ; 6(2): 215-224, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32313680

ABSTRACT

OBJECTIVE: There is increasing evidence that immune cell interactions in adipose tissue contribute to the development of metabolic dysfunction. Pro-inflammatory cytokines have been shown to mediate insulin resistance, and the presence of macrophages is a salient feature in the development of obesity. The present study aimed to evaluate adipocyte size and macrophage activation in women before and 3 months after laparoscopic vertical sleeve gastrectomy (VSG). METHODS: Subcutaneous abdominal adipose tissue biopsies were obtained from women scheduled to undergo VSG. Histological evaluation of adipocytes and macrophages was performed as well as cytokine expression quantification before and after VSG-induced weight loss. RESULTS: Weight loss following VSG resulted in a reduction in adipocyte size as well as a decrease in interleukin (IL)-6 cytokine mRNA expression in subcutaneous adipose tissue. There was no change in the presence of crownlike structures after weight loss. CONCLUSIONS: Early weight loss after VSG is associated with a reduction in adipocyte size and a decline in IL-6 gene expression in local adipose tissue. Macrophage infiltration and crownlike density structures persist in adipose tissue from tissues impacted by excess body weight 3 months after VSG-induced weight loss.

10.
Sci Rep ; 10(1): 1560, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005914

ABSTRACT

BACKGROUND: Intrinsically photosensitive retinal ganglion cells (ipRGCs) control non-visual light responses (e.g. pupillary light reflex and circadian entrainment). Patients with diabetic retinopathy (DR) show reduced ipRGC function, as inferred by abnormalities in the post illumination pupil response (PIPR). We explored whether ipRGC function in DR is associated with circadian outputs and sleep/wake behavior. METHODS: Forty-five participants (15 without diabetes, 15 with type 2 diabetes (T2D) and no DR, 15 with T2D and DR) participated. ipRGC function was inferred from the PIPR (pupil size following stimulus offset). Circadian outputs were melatonin amplitude (overnight urinary 6-sulfatoxymelatonin (aMT6s)) and timing (dim light melatonin onset (DLMO)), and evening salivary cortisol levels. Sleep/wake patterns were measured with wrist actigraphy and insomnia symptoms were assessed subjectively. RESULTS: Patients with T2D and DR had smaller PIPR and lower urinary aMT6s than other groups (p < 0.001). In adjusted regression models, smaller PIPR was associated with lower urinary aMT6s (ß = 4.552, p = 0.005). Patients with DR were more likely to have no detectable DLMO (p = 0.049), higher evening salivary cortisol, greater insomnia symptoms and greater sleep variability compared to other groups. Sleep duration, efficiency and rest-activity rhythms were similar. CONCLUSION: Reduced ipRGC function in DR is associated with circadian dysregulation and sleep disturbances, although a causal relationship cannot be established in this cross-sectional study. Prospective mechanistic and intervention studies examining circadian and sleep health in these patients are warranted.


Subject(s)
Adie Syndrome/metabolism , Circadian Clocks/physiology , Diabetes Mellitus, Type 2/metabolism , Diabetic Retinopathy/metabolism , Retinal Ganglion Cells/physiology , Sleep Disorders, Circadian Rhythm/metabolism , Adie Syndrome/pathology , Aged , Cells, Cultured , Cross-Sectional Studies , Diabetes Mellitus, Type 2/pathology , Diabetic Retinopathy/pathology , Female , Humans , Hydrocortisone/metabolism , Male , Melatonin/analogs & derivatives , Melatonin/metabolism , Melatonin/urine , Middle Aged , Reflex, Pupillary , Sleep Disorders, Circadian Rhythm/pathology , Sleep Initiation and Maintenance Disorders
11.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-31970413

ABSTRACT

CONTEXT: The endocannabinoid (eCB) system partly controls hedonic eating, a major cause of obesity. While some studies suggested an overactivation of the eCB system in obesity, peripheral levels of eCBs across the 24-hour cycle have not been characterized in obese individuals despite the fact that in lean adults, levels of the eCB 2-arachidonoylglycerol (2-AG) vary across the day. OBJECTIVE: We sought to examine 24-hour profiles of serum concentrations of 2-AG in healthy obese and nonobese adults, under well-controlled laboratory conditions. We also simultaneously assessed 24-hour profiles of 2-oleoylglycerol (2-OG), leptin, and cortisol in each participant. DESIGN: With fixed light-dark and sleep-wake cycles, blood sampling was performed over an entire 24-hour period, including identical meals at 0900, 1400, and 1900. PARTICIPANTS: Twelve obese (8 women, mean body mass index [BMI]: 39.1 kg/m2) and 15 nonobese (6 women; mean BMI: 23.6 kg/m2) healthy adults were studied. RESULTS: We observed a 24-hour variation of 2-AG levels in obese individuals but, relative to nonobese adults, the amplitude was dampened and the timings of the nadir and peak were delayed by 4 to 5 hours. The profile of 2-OG was similarly misaligned. In contrast, when expressed relative to the 24-hour mean level, the 24-hour rhythm of cortisol and leptin were similar in obese and nonobese participants. CONCLUSIONS: Obesity appears to be associated with a dampening and delay of the 24-hour variation of eCB activity relative to the central circadian signal as well as to the daily leptin rhythm. This misalignment may play a role in the pathophysiology of obesity.


Subject(s)
Arachidonic Acids/blood , Circadian Rhythm/physiology , Endocannabinoids/blood , Glycerides/blood , Obesity/blood , Adult , Case-Control Studies , Female , Humans , Hydrocortisone/blood , Leptin/blood , Male , Obesity/physiopathology , Young Adult
12.
Psychoneuroendocrinology ; 111: 104471, 2020 01.
Article in English | MEDLINE | ID: mdl-31610409

ABSTRACT

OBJECTIVE: The endocannabinoid (eCB) system is involved in diverse aspects of human physiology and behavior but little is known about the impact of circadian rhythmicity on the system. The two most studied endocannabinoids, AEA (ananamide) and 2-AG (2-arachidonoylglycerol), can be measured in peripheral blood however the functional relevance of peripheral eCB levels is not clear. Having previously detailed the 24-h profile of serum 2-AG, here we report the 24-h serum profile of AEA to determine if these two endocannabinoids vary in parallel across the biological day including a nocturnal 8.5-h sleep period. Further, we assessed and compared the effect of a physiological challenge, in the form of sleep restriction to 4.5-h, on these two profiles. METHODS: In this randomized crossover study, we examined serum concentrations of AEA across a 24-h period in fourteen young adults. Congeners of AEA, the structural analogs oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) were simultaneously assayed. Prior to 24-h blood sampling, each participant was exposed to two nights of normal (8.5 h) or restricted sleep (4.5 h). The two sleep conditions were separated by at least one month. In both sleep conditions, during the period of blood sampling, each individual ate the same high-carbohydrate meal at 0900, 1400, and 1900. RESULTS: Mean 24-h concentrations of AEA were 0.697 ±â€¯0.11 pmol/ml. A reproducible biphasic 24-h profile of AEA was observed with a first peak occurring during early sleep (0200) and a second peak in the mid-afternoon (1500) while a nadir was detected in the mid-morning (1000). The 24-h profiles for both OEA and PEA followed a similar pattern to that observed for AEA. AEA, OEA, and PEA levels were not affected by sleep restriction at any time of day, contrasting with the elevation of early afternoon levels previously observed for 2-AG. CONCLUSIONS: The 24-h rhythm of AEA is markedly different from that of 2-AG, being of lesser amplitude and biphasic, rather than monophasic. These observations suggest distinct regulatory pathways of the two eCB and indicate that time of day needs to be carefully controlled in studies attempting to delineate their relative roles. Moreover, unlike 2-AG, AEA is not altered by sleep restriction, suggesting that physiological perturbations may affect AEA and 2-AG differently. Similar 24-h profiles were observed for OEA and PEA following normal and restricted sleep, further corroborating the validity of the wave-shape and lack of response to sleep loss observed for the AEA profile. Therapeutic approaches involving agonism or antagonism of peripheral eCB signaling will likely need to be tailored according to time of day.


Subject(s)
Arachidonic Acids/metabolism , Circadian Rhythm/physiology , Endocannabinoids/metabolism , Glycerides/metabolism , Adolescent , Adult , Amides , Arachidonic Acids/blood , Arachidonic Acids/physiology , Cross-Over Studies , Endocannabinoids/analysis , Endocannabinoids/blood , Endocannabinoids/physiology , Ethanolamines/analysis , Ethanolamines/blood , Female , Glycerides/blood , Glycerides/physiology , Humans , Male , Oleic Acids/analysis , Oleic Acids/blood , Palmitic Acids/analysis , Palmitic Acids/blood , Polyunsaturated Alkamides , Sleep/physiology , Young Adult
13.
J Biol Rhythms ; 35(1): 84-97, 2020 02.
Article in English | MEDLINE | ID: mdl-31668115

ABSTRACT

A hallmark of biology is the cyclical nature of organismal physiology driven by networks of biological, including circadian, rhythms. Unsurprisingly, disruptions of the circadian rhythms through sleep curtailment or shift work have been connected through numerous studies to positive associations with obesity, insulin resistance, and diabetes. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) measures oscillation in messenger RNA expression, an essential foundation for the study of the physiological circadian regulatory network. Primarily, measured oscillations have involved the use of reference gene normalization. However, the validation and identification of suitable reference genes is a significant challenge across different biological systems. This study focuses on adipose tissue of premenopausal, otherwise healthy, morbidly obese women voluntarily enrolled after being scheduled for laparoscopic sleeve gastrectomy surgery. Acquisition of tissue was accomplished by aspiratory needle biopsies of subcutaneous adipose tissue 1 to 2 weeks prior to surgery and 12 to 13 weeks following surgery and an in-surgery scalpel-assisted excision of mesenteric adipose tissue. Each biopsy was sterile cultured ex vivo and serially collected every 4 h over approximately 36 h. The candidate reference genes that were tested were 18S rRNA, GAPDH, HPRT1, RPII, RPL13α, and YWHAZ. Three analytic tools were used to test suitability, and the candidate reference genes were used to measure oscillation in expression of a known circadian clock element (Dbp). No gene was deemed suitable as an individual reference gene control, which indicated that the optimal reference gene set was the geometrically averaged 3-gene panel composed of YWHAZ, RPL13α, and GAPDH. These methods can be employed to identify optimal reference genes in other systems.


Subject(s)
Adipose Tissue , Circadian Rhythm/genetics , Gene Expression Profiling/standards , Gene Expression , Real-Time Polymerase Chain Reaction/standards , Adult , Circadian Rhythm/physiology , Female , Gene Expression Profiling/methods , Humans , Middle Aged , Obesity, Morbid , Real-Time Polymerase Chain Reaction/methods , Reference Standards , Young Adult
14.
Am J Physiol Regul Integr Comp Physiol ; 311(3): R558-63, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27465735

ABSTRACT

Thirteen percent of the world's population suffers from obesity and 39% from being overweight, which correlates with an increase in numerous secondary metabolic complications, such as Type 2 diabetes mellitus. Bariatric surgery is the most effective treatment for severe obesity and results in significant weight loss and the amelioration of obesity-related comorbidities through changes in enteroendocrine activity, caloric intake, and alterations in gut microbiota composition. The circadian system has recently been found to be a critical regulatory component in the control of metabolism and, thus, may potentially play an important role in inappropriate weight gain. Indeed, some behaviors and lifestyle factors associated with an increased risk of obesity are also risk factors for misalignment in the circadian clock system and for the metabolic syndrome. It is thus possible that alterations in peripheral circadian clocks in metabolically relevant tissues are a contributor to the current obesity epidemic. As such, it is plausible that postsurgical alterations in central circadian alignment, as well as peripheral gene expression in metabolic tissues may represent another mechanism for the beneficial effects of bariatric surgery. Bariatric surgery may represent an opportunity to identify changes in the circadian expression of clock genes that have been altered by environmental factors, allowing for a better understanding of the mechanism of action of surgery. These studies could also reveal an overlooked target for behavioral intervention to improve metabolic outcomes following bariatric surgery.


Subject(s)
Bariatric Surgery , Circadian Rhythm , Insulin Resistance , Models, Biological , Obesity/physiopathology , Obesity/surgery , CLOCK Proteins/metabolism , Humans , Prognosis , Treatment Outcome
15.
Sleep ; 39(3): 653-64, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26612385

ABSTRACT

STUDY OBJECTIVES: Increasing evidence from laboratory and epidemiologic studies indicates that insufficient sleep may be a risk factor for obesity. Sleep curtailment results in stimulation of hunger and food intake that exceeds the energy cost of extended wakefulness, suggesting the involvement of reward mechanisms. The current study tested the hypothesis that sleep restriction is associated with activation of the endocannabinoid (eCB) system, a key component of hedonic pathways involved in modulating appetite and food intake. METHODS: In a randomized crossover study comparing 4 nights of normal (8.5 h) versus restricted sleep (4.5 h) in healthy young adults, we examined the 24-h profiles of circulating concentrations of the endocannabinoid 2-arachidonoylglycerol (2-AG) and its structural analog 2-oleoylglycerol (2-OG). We concomitantly assessed hunger, appetite, and food intake under controlled conditions. RESULTS: A robust daily variation of 2-AG concentrations with a nadir around the middle of the sleep/overnight fast, followed by a continuous increase culminating in the early afternoon, was evident under both sleep conditions but sleep restriction resulted in an amplification of this rhythm with delayed and extended maximum values. Concentrations of 2-OG followed a similar pattern, but with a lesser amplitude. When sleep deprived, participants reported increases in hunger and appetite concomitant with the afternoon elevation of 2-AG concentrations, and were less able to inhibit intake of palatable snacks. CONCLUSIONS: Our findings suggest that activation of the eCB system may be involved in excessive food intake in a state of sleep debt and contribute to the increased risk of obesity associated with insufficient sleep. COMMENTARY: A commentary on this article appears in this issue on page 495.


Subject(s)
Arachidonic Acids/blood , Circadian Rhythm/physiology , Endocannabinoids/blood , Glycerides/blood , Hyperphagia/blood , Hyperphagia/physiopathology , Sleep Deprivation/blood , Sleep Deprivation/physiopathology , Adolescent , Adult , Appetite Regulation/physiology , Cross-Over Studies , Eating/physiology , Fasting/blood , Female , Healthy Volunteers , Humans , Hunger/physiology , Hyperphagia/etiology , Male , Obesity/blood , Obesity/etiology , Reward , Sleep/physiology , Sleep Deprivation/complications , Wakefulness , Young Adult
16.
Sleep ; 38(12): 1849-60, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26564131

ABSTRACT

A workshop was held at the National Institute for Diabetes and Digestive and Kidney Diseases with a focus on the impact of sleep and circadian disruption on energy balance and diabetes. The workshop identified a number of key principles for research in this area and a number of specific opportunities. Studies in this area would be facilitated by active collaboration between investigators in sleep/circadian research and investigators in metabolism/diabetes. There is a need to translate the elegant findings from basic research into improving the metabolic health of the American public. There is also a need for investigators studying the impact of sleep/circadian disruption in humans to move beyond measurements of insulin and glucose and conduct more in-depth phenotyping. There is also a need for the assessments of sleep and circadian rhythms as well as assessments for sleep-disordered breathing to be incorporated into all ongoing cohort studies related to diabetes risk. Studies in humans need to complement the elegant short-term laboratory-based human studies of simulated short sleep and shift work etc. with studies in subjects in the general population with these disorders. It is conceivable that chronic adaptations occur, and if so, the mechanisms by which they occur needs to be identified and understood. Particular areas of opportunity that are ready for translation are studies to address whether CPAP treatment of patients with pre-diabetes and obstructive sleep apnea (OSA) prevents or delays the onset of diabetes and whether temporal restricted feeding has the same impact on obesity rates in humans as it does in mice.


Subject(s)
Circadian Rhythm/physiology , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Energy Metabolism , Sleep Disorders, Circadian Rhythm/physiopathology , Sleep/physiology , Age of Onset , Animals , Diabetes Mellitus/etiology , Endophenotypes , Fasting/metabolism , Food Deprivation , Glucose/metabolism , Homeostasis , Humans , Insulin/metabolism , Metabolic Syndrome/metabolism , Mice , Obesity/metabolism , Obesity/physiopathology , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/therapy , Sleep Disorders, Circadian Rhythm/metabolism , Time Factors
17.
Neuropsychopharmacology ; 38(13): 2578-87, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23864029

ABSTRACT

Epidemiological studies have shown a link between sleep loss and the obesity 'epidemic,' and several observations indicate that sleep curtailment engenders positive energy balance via increased palatable-food 'snacking.' These effects suggest alterations in reward-modulatory brain systems. We explored the effects of 10 days of sleep deprivation in rats on the expression of striatal opioid peptide (OP) genes that subserve food motivation and hedonic reward, and compared effects with those seen in hypothalamic energy balance-regulatory systems. Sleep-deprived (Sleep-Dep) rats were compared with yoked forced-locomotion apparatus controls (App-Controls), food-restricted rats (Food-Restrict), and unmanipulated controls (Home-Cage). Detection of mRNA levels with in situ hybridization revealed a subregion-specific upregulation of striatal preproenkephalin and prodynorhin gene expression in the Sleep-Dep group relative to all other groups. Neuropeptide Y (NPY) gene expression in the hippocampal dentate gyrus and throughout neocortex was also robustly upregulated selectively in the Sleep-Dep group. In contrast, parallel gene expression changes were observed in the Sleep-Dep and Food-Restrict groups in hypothalamic energy-sensing systems (arcuate nucleus NPY was upregulated, and cocaine- and amphetamine-regulated transcript was downregulated), in alignment with leptin suppression in both groups. Together, these results reveal a novel set of sleep deprivation-induced transcriptional changes in reward-modulatory peptide systems, which are dissociable from the energy-balance perturbations of sleep loss or the potentially stressful effects of the forced-locomotion procedure. The recruitment of telencephalic food-reward systems may provide a feeding drive highly resistant to feedback control, which could engender obesity through the enhancement of palatable feeding.


Subject(s)
Neostriatum/metabolism , Opioid Peptides/metabolism , Reward , Sleep Deprivation/pathology , Up-Regulation/physiology , Animals , Body Weight , Eating , Enkephalins/genetics , Enkephalins/metabolism , Fasting/physiology , Insulin/blood , Leptin/blood , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroimaging , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Opioid Peptides/genetics , Protein Precursors/genetics , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley
18.
Neurosci Biobehav Rev ; 37(9 Pt A): 1985-98, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23466532

ABSTRACT

Circuits that participate in specific subcomponents of feeding (e.g., gustatory perception, peripheral feedback relevant to satiety and energy balance, reward coding, etc.) are found at all levels of the neural axis. Further complexity is conferred by the wide variety of feeding-modulatory neurotransmitters and neuropeptides that act within these circuits. An ongoing challenge has been to refine the understanding of the functional specificity of these neurotransmitters and circuits, and there have been exciting advances in recent years. We focus here on foundational work of Dr. Ann Kelley that identified distinguishable actions of striatal opioid peptide modulation and dopamine transmission in subcomponents of reward processing. We also discuss her work in overlaying these neuropharmacological effects upon anatomical pathways that link the telencephalon (cortex and basal ganglia) with feeding-control circuits in the hypothalamus. Using these seminal contributions as a starting point, we will discuss new findings that expand our understanding of (1) the specific, differentiable motivational processes that are governed by central dopamine and opioid transmission, (2) the manner in which other striatal neuromodulators, specifically acetylcholine, endocannabinoids and adenosine, modulate these motivational processes (including via interactions with opioid systems), and (3) the organization of the cortical-subcortical network that subserves opioid-driven feeding. The findings discussed here strengthen the view that incentive-motivational properties of food are coded by substrates and neural circuits that are distinguishable from those that mediate the acute hedonic experience of food reward. Striatal opioid transmission modulates reward processing by engaging frontotemporal circuits, possibly via a hypothalamic-thalamic axis, that ultimately impinges upon hypothalamic modules dedicated to autonomic function and motor pattern control. We will conclude by discussing implications for understanding disorders of "non-homeostatic" feeding.


Subject(s)
Feeding Behavior/physiology , Motivation/physiology , Reward , Animals , Feeding Behavior/drug effects , Humans , Motivation/drug effects , Neural Pathways , Neuroanatomy , Neuropharmacology
19.
Diabetes Care ; 35(11): 2384-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22912423

ABSTRACT

OBJECTIVE: To assess whether the presence of obstructive sleep apnea (OSA) affects glucose metabolism in young, lean individuals who are healthy and free of cardiometabolic disease. RESEARCH DESIGN AND METHODS: In a prospective design, 52 healthy men (age 18-30 years; BMI 18-25 kg/m(2)) underwent laboratory polysomnogram followed by a morning oral glucose tolerance test (OGTT). We stratified all subjects according to the presence or absence of ethnicity-based diabetes risk and family history of diabetes. We then used a frequency-matching approach and randomly selected individuals without OSA, yielding a total of 20 control men without OSA and 12 men with OSA. Indices of glucose tolerance, insulin sensitivity, and insulin secretion (early phase and total) were compared between men with OSA and control subjects. The incremental areas under the glucose (incAUC(glu)) and insulin (incAUC(ins)) curves were calculated using the trapezoidal method from 0 to 120 min during the OGTT. RESULTS: Men with OSA and control subjects were similar in terms of age, BMI, ethnicity-based diabetes risk, family history of diabetes, and level of exercise. Both groups had normal systolic and diastolic blood pressure and fasting lipid levels. After ingestion of a glucose load, men with OSA had 27% lower insulin sensitivity (estimated by Matsuda index) and 37% higher total insulin secretion (incAUC(ins)) than the control subjects, despite comparable glucose levels (incAUC(glu)). CONCLUSIONS: In young, lean, and healthy men who are free of cardiometabolic disease, the presence of OSA is associated with insulin resistance and a compensatory rise in insulin secretion to maintain normal glucose tolerance. Thus, OSA may increase the risk of type 2 diabetes independently of traditional cardiometabolic risk factors.


Subject(s)
Insulin Resistance/physiology , Insulin/metabolism , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/physiopathology , Thinness/metabolism , Adolescent , Adult , Humans , Insulin Secretion , Male , Prospective Studies , Young Adult
20.
Curr Top Med Chem ; 11(19): 2472-82, 2011.
Article in English | MEDLINE | ID: mdl-21906017

ABSTRACT

Sleep is homeostatically regulated in all species that have been carefully studied. In mammals and birds, the best characterized marker of sleep pressure is slow wave activity (SWA), defined as the electroencephalogram (EEG) power between 0.5 and 4 Hz during NREM sleep. SWA peaks at sleep onset and decreases with time spent asleep, and reflects the synchronous firing of cortical neurons coordinated by an underlying slow oscillation, the fundamental cellular phenomenon of NREM sleep. We have recently proposed the synaptic homeostasis hypothesis of sleep, which claims that an important function of sleep is to maintain synaptic balance. This hypothesis states that plastic processes during wake are biased towards synaptic potentiation, resulting in a net increase in synaptic strength in many brain circuits. Such increased synaptic weight would be unsustainable in the long run, due to increased demand for energy, space and supplies, and risk of synaptic saturation. Thus, according to the synaptic homeostasis hypothesis, sleep is important to renormalize synaptic strength to a baseline level that is sustainable and beneficial for memory and performance. There is strong evidence that the amplitude and slope of EEG slow waves is related to the number of neurons that enter an up state or a down state of the slow oscillation near-synchronously, and that synchrony is directly related to the number, strength, and efficacy of synaptic connections among them. Thus, the average synaptic strength (number or efficacy of synapses) reached in a given cortical area at the end of the major wake phase should be reflected by the level of SWA in the EEG at sleep onset. Moreover, according to the hypothesis, sleep SWA is not only a useful proxy of wake-related cortical synaptic strength, but could mediate the renormalization of neural circuits by favoring net synaptic depression, perhaps aided by low levels of norepinephrine, serotonin, and acetylcholine during NREM sleep. Here we briefly review human and animal studies showing that, consistent with this hypothesis, 1) in the adult cerebral cortex wake is associated with a net increase in synaptic strength, and sleep with a net decrease; and 2) SWA reflects not just prior "use" of specific neuronal circuits, but rather the occurrence of plastic changes, with increases in SWA after synaptic potentiation, and decreases in SWA after synaptic depression. We end by discussing current challenges to this hypothesis and future research directions.


Subject(s)
Sleep/physiology , Synapses/physiology , Animals , Electroencephalography , Electrophysiological Phenomena , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...