1
|
Ashcroft SP, Ehrlich AM, Burek K, Pendergrast LA, Yonamine CY, Treebak JT, Zierath JR. Enhanced metabolic adaptations following late dark phase wheel running in high-fat diet-fed mice. Mol Metab 2025; 95:102116. [PMID: 39993626 PMCID: PMC11930447 DOI: 10.1016/j.molmet.2025.102116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025] Open
Abstract
Exercise interventions represent an effective strategy to prevent and treat metabolic diseases and the time-of-day-dependent effects of exercise on metabolic outcomes are becoming increasingly apparent. We aimed to study the influence of time-restricted wheel running on whole-body energy and glucose homeostasis. Male, 8-week-old, C57BL/6NTac mice were fed either a 60% high-fat diet (HFD) or a 10% low-fat diet (LFD) for 4 weeks. Following this, mice were given access to a running wheel between zeitgeber time (ZT) 12-16 (early dark phase) or ZT 20-0 (late dark phase). Sedentary mice had access to a permanently locked wheel. Mice were housed under these conditions in metabolic chambers for 4 weeks in which LFD and HFD conditions were maintained. Following the exercise intervention, body composition and glucose tolerance were assessed. Wheel running during either the early or late dark phase resulted in metabolic improvements such as attenuation in body weight gain, enhanced glucose tolerance and reduced ectopic lipid deposition. However, late dark phase exercise resulted in a greater reduction in body weight gain, as well as enhanced metabolic flexibility and insulin sensitivity. Our data suggest that late dark phase versus early dark phase exercise confers greater metabolic adaptations in HFD-fed mice.
Collapse
Affiliation(s)
- Stephen P Ashcroft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Krzysztof Burek
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Logan A Pendergrast
- Integrative Physiology Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Caio Y Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Integrative Physiology Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Integrative Physiology Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Yang Y, Guo L, Li M, Li Z, Xiao Y, Wang Y, Li Y, Deng J, Wu S, Zhang H. Joint association of physical activity and night sleep duration with the risk of atherosclerotic cardiovascular disease. J Stroke Cerebrovasc Dis 2025; 34:108258. [PMID: 39921196 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVES To investigate the joint association of physical activity (PA) and sleep duration with the risk of atherosclerotic cardiovascular disease (ASCVD). METHODS A total of 111,714 participants who participated in health examinations were enrolled from the Kailuan Study between June 2006 and December 2009. Sleep duration was divided into three groups (short, healthy, and long). PA was categorized into physically inactive and physically active. Restricted cubic splines were used to analyze the associations of PA and sleep duration with ASCVD. Cox proportional hazards models were conducted to investigate the effects of PA and sleep duration on ASCVD risk, with their cross-product interaction terms tested on both multiplicative and additive scales. RESULTS During 13.05 ± 3.16 years of follow-up, 10,408 participants developed ASCVD. Restricted cubic spline analysis revealed a nonlinear dose-response relationship between sleep duration and ASCVD risk among physically inactive participants (Pnonlinear <0.05). Both additive and multiplicative interactions between physical activity and sleep duration were observed in their effects on ASCVD risk (Pinteraction < 0.05). Compared with participants who had healthy sleep duration and were physically active, those with unhealthy sleep duration and physical inactivity exhibited a significantly higher risk of ASCVD [short sleep duration: HR (95 %CI) = 1.19 (1.09, 1.30); long sleep duration: HR (95 % CI) = 1.10 (0.90, 1.34)]. However, physically active participants in both the short and long sleep duration groups exhibited a reduced risk of ASCVD. CONCLUSIONS Engaging in sufficient PA can reduce the risk of ASCVD in individuals with short sleep duration. Therefore, simultaneously maintaining healthy sleep duration and engaging in PA may effectively prevent ASCVD.
Collapse
Affiliation(s)
- Yuxin Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Lu Guo
- School of Public Health North China University of Science and Technology, Tangshan, Hebei, China.
| | - Man Li
- Graduate School, North China University of Science and Technology, Tangshan 063000, China.
| | - Zhiqiang Li
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yao Xiao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yuanyuan Li
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Jiawen Deng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China.
| | - Hong Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Yin X, Bao W, Ley SH, Yang J, Cuffe SB, Yu G, Chavarro JE, Liu P, Zhou JH, Tobias DK, Hu FB, Zhang C. Sleep Characteristics and Long-Term Risk of Type 2 Diabetes Among Women With Gestational Diabetes. JAMA Netw Open 2025; 8:e250142. [PMID: 40042841 PMCID: PMC11883505 DOI: 10.1001/jamanetworkopen.2025.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/26/2024] [Indexed: 03/09/2025] Open
Abstract
Importance Women with a history of gestational diabetes (GD) are at high risk for developing type 2 diabetes (T2D). Sleep is a crucial lifestyle factor associated with cardiometabolic health, yet studies on its role in the progression from GD to T2D are sparse. Objective To investigate the associations of sleep duration and quality with T2D risk and levels of glucose metabolism biomarkers in women with a history of GD. Design, Setting, and Participants This cohort study used data from the Nurses' Health Study II, an ongoing longitudinal cohort that began in 1989 and initially included 116 429 female nurses with health status and lifestyle factors updated every 2 to 4 years. A subset of participants with a history of GD was followed up through June 2021. Sleep characteristics were assessed in the 2001 questionnaire (administered from June 2001 to June 2003), which served as the baseline for follow-up. Data were analyzed from November 2023 to August 2024. Exposures Snoring frequency, sleep duration, and daytime sleepiness. Main Outcomes and Measures Physician-diagnosed incident T2D was ascertained biennially via questionnaires. Cox proportional hazards regression models were used to estimate the hazard ratios (HRs) and 95% CIs. Biomarkers (glycated hemoglobin [HbA1c], C-peptide, and insulin) were compared across sleep characteristic categories using multivariable-adjusted least-squares means (LSMs) and 95% CIs. Results During a total of 42 155 person-years of follow-up among 2891 women with a history of GD (mean [SD] age, 45.3 [4.4] years), 563 women (19.5%) developed T2D. Compared with women who reported rarely snoring, those with occasional or regular snoring had significantly higher T2D risk, with adjusted HRs of 1.54 (95% CI, 1.18-2.02) and 1.61 (95% CI, 1.21-2.13), respectively. Compared with women who slept 7 to 8 hours per day, shorter sleep duration (≤6 hours per day) was significantly associated with a higher risk of T2D (HR, 1.32; 95% CI, 1.06-1.64). Women who slept 6 or fewer hours per day and snored regularly had the highest risk of developing T2D (HR, 2.06; 95% CI, 1.38-3.07). Additionally, more frequent snoring was associated with higher HbA1c (LSM, 5.89; 95% CI, 5.75-6.02), C-peptide (LSM, 4.30; 95% CI, 3.70-4.99), and insulin (LSM, 11.25; 95% CI, 8.75-14.40) levels in the full adjusted models (P = .01 for trend for all). Conclusions and Relevance In this cohort study of women with a history of GD, shorter sleep duration and both occasional and regular snoring were significantly associated with an increased risk of T2D. These findings suggest that improving sleep health may be important to reduce T2D incidence in this high-risk population.
Collapse
Affiliation(s)
- Xin Yin
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei Bao
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Sylvia H. Ley
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Jiaxi Yang
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sherri Babaeian Cuffe
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Guoqi Yu
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jorge E. Chavarro
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peipei Liu
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Medicine & Human Potential Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Deirdre K. Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Division of Preventive Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Frank B. Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Cuilin Zhang
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
4
|
Apostolopoulou M, Lambadiari V, Roden M, Dimitriadis GD. Insulin Resistance in Type 1 Diabetes: Pathophysiological, Clinical, and Therapeutic Relevance. Endocr Rev 2025:bnae032. [PMID: 39998445 DOI: 10.1210/endrev/bnae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Indexed: 02/26/2025]
Abstract
People with type 1 diabetes (T1D) are usually considered to exclusively exhibit β-cell failure, but they frequently also feature insulin resistance. This review discusses the mechanisms, clinical features, and therapeutic relevance of insulin resistance by focusing mainly on human studies using gold-standard techniques (euglycemic-hyperinsulinemic clamp). In T1D, tissue-specific insulin resistance can develop early and sustain throughout disease progression. The underlying pathophysiology is complex, involving both metabolic- and autoimmune-related factors operating synergistically. Insulin treatment may play an important pathogenic role in predisposing individuals with T1D to insulin resistance. However, the established lifestyle-related risk factors and peripheral insulin administration inducing glucolipotoxicity, hyperinsulinemia, hyperglucagonemia, inflammation, mitochondrial abnormalities, and oxidative stress cannot always fully explain insulin resistance in T1D, suggesting a phenotype distinct from type 2 diabetes. The mutual interaction between insulin resistance and impaired endothelial function further contributes to diabetes-related complications. Insulin resistance should therefore be considered a treatment target in T1D. Aside from lifestyle modifications, continuous subcutaneous insulin infusion can ameliorate insulin resistance and hyperinsulinemia, thereby improving glucose toxicity compared with multiple injection insulin treatment. Among other concepts, metformin, pioglitazone, incretin-based drugs such as GLP-1 receptor agonists, sodium-glucose cotransporter inhibitors, and pramlintide can improve insulin resistance, either directly or indirectly. However, considering the current issues of high cost, side effects, limited efficacy, and their off-label status, these agents in people with T1D are not widely used in routine clinical care at present.
Collapse
Affiliation(s)
- Maria Apostolopoulou
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibnitz Center for Diabetes Research at Heinrich-Heine University, 40225 Düsseldorf, Germany
- German Center of Diabetes Research (DZD), Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibnitz Center for Diabetes Research at Heinrich-Heine University, 40225 Düsseldorf, Germany
- German Center of Diabetes Research (DZD), Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - George D Dimitriadis
- 2nd Department of Internal Medicine, Research Institute and Diabetes Center, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece
| |
Collapse
|
5
|
Lawson Y, Wilding GE, El-Solh AA. Insomnia and risk of mortality in older adults. J Sleep Res 2024; 33:e14229. [PMID: 38685752 DOI: 10.1111/jsr.14229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
Existing evidence linking insomnia to all-cause mortality in older individuals remains inconclusive. We conducted a retrospective study of a large cohort of veterans aged 65-80 years old identified from the Corporate Data Warehouse, a large data repository derived from the Veterans Health Administration integrated medical records. Veterans' enrollees with and without International Classification of Diseases, Ninth and Tenth Revision, codes corresponding to insomnia diagnosis between 1 January 2010 and 30 March 2019 were assessed for eligibility. The primary outcome was all-cause mortality. A total of 36,269 veterans, 9584 with insomnia and 26,685 without insomnia, were included in the analysis. Baseline mean (SD) age was 72.6 (4.2) years. During a mean follow-up of 6.0 (2.9) years of the propensity score matched sample, the mortality rate was 34.8 [95% confidence interval: 33.2-36.6] deaths per 1000 person-years among patients with insomnia compared with 27.8 [95% confidence interval: 26.6-29.1] among patients without insomnia. In a Cox proportional hazards model, insomnia was significantly associated with higher mortality (hazard ratio: 1.39; [95% confidence interval: 1.27-1.52]). Patients with insomnia also had a higher risk of non-fatal cardiovascular events (hazard ratio: 1.21; [95% confidence interval: 1.06-1.37]). Secondary stratified analyses by sex, race, ethnicity and hypertension showed no evidence of effect modification. A higher risk of mortality (hazard ratio: 1.51; [95% confidence interval: 1.33-1.71]) was observed when depression was present compared with absent (hazard ratio: 1.26; [95% confidence interval: 1.12-1.44]; p = 0.02). In this cohort study, insomnia was associated with increased risk-adjusted mortality and non-fatal cardiovascular events in older individuals.
Collapse
Affiliation(s)
- Yolanda Lawson
- The Veterans Affairs Western New York Healthcare System, Buffalo, New York, USA
| | - Gregory E Wilding
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Ali A El-Solh
- The Veterans Affairs Western New York Healthcare System, Buffalo, New York, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Jacobs School of Medicine, Buffalo, New York, USA
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
6
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
7
|
Lin J, Song Z, Li Y, Chiang C, Hirakawa Y, Nakano Y, Hong YJ, Matsunaga M, Ota A, Tamakoshi K, Yatsuya H. Nonrestorative Sleep and Type 2 Diabetes Incidence: The Aichi Workers' Cohort Study. J Epidemiol 2024; 34:428-433. [PMID: 38281747 PMCID: PMC11330709 DOI: 10.2188/jea.je20230184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/08/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND The term "nonrestorative sleep (NRS)" refers to an unrefreshed feeling at wake-up and is a domain of poor sleep quality. Previous research has demonstrated that NRS is linked to a number of diseases and adverse health outcomes, but less is known regarding the link between NRS and diabetes, particularly in Japanese. METHODS We studied 3,665 middle-aged male participants of the Aichi Workers' Cohort Study who were followed-up from 2002 through 2019. Cox proportional hazards models estimated hazard ratios (HRs) and 95% confidence intervals (CIs) of incident type 2 diabetes mellitus (T2DM) in relation to NRS adjusted for potential confounding variables. RESULTS During a median follow-up of 14.6 years, 421 type 2 diabetes cases were identified. Participants with NRS had a higher crude incidence rate of T2DM (11.2/1,000 person-years), compared to participants without NRS (9.3/1,000 person-years). In the fully adjusted model, individuals who reported having NRS had a significantly higher risk of developing T2DM (HR1.36; 95% CI, 1.10-1.67). The association was observed only in participants under 50 years old (HR 1.82; 95% CI, 1.36-2.43), not in the older (50 years or older) participants (P for interaction = 0.025). In contrast, stratified analyses by the presence of shift work, obesity, or sleep duration showed similar associations in all the strata. CONCLUSION NRS was associated with higher risk of T2DM in middle-aged Japanese male workers independent of a variety of lifestyle factors and other sleep problems.
Collapse
Affiliation(s)
- Jingyi Lin
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Zean Song
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuanying Li
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chifa Chiang
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Hirakawa
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Nakano
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Young-Jae Hong
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaaki Matsunaga
- Department of Public Health, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Atsuhiko Ota
- Department of Public Health, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Koji Tamakoshi
- Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Yatsuya
- Department of Public Health and Health System, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
8
|
Souza JFT, Monico-Neto M, Tufik S, Antunes HKM. Sleep Debt and Insulin Resistance: What's Worse, Sleep Deprivation or Sleep Restriction? Sleep Sci 2024; 17:e272-e280. [PMID: 39268336 PMCID: PMC11390169 DOI: 10.1055/s-0044-1782173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 10/05/2023] [Indexed: 09/15/2024] Open
Abstract
Objective To evaluate which condition of sleep debt has a greater negative impact on insulin resistance: sleep deprivation for 24 hours or 4 hours of sleep restriction for 4 nights. Materials and Methods In total, 28 healthy male subjects aged 18 to 40 years were recruited and randomly allocated to two groups: sleep deprivation (SD) and sleep restriction (SR). Each group underwent two conditions: regular sleep (11 pm to 7 am ) and total sleep deprivation for 24 hours (SD); regular sleep (11 pm to 7 am ) and 4 nights of sleep restriction (SR) (1 am to 5 am ). The oral glucose tolerance test (OGTT) was performed, and baseline glucose, insulin, free fatty acids (FFAs), and cortisol were measured. In addition, the area under the curve (AUC) for glucose and insulin, the homeostasis model assessment of insulin resistance (HOMA-IR), and the Matsuda Index (Insulin Sensitivity Index, ISI) were calculated. Results Glucose and insulin had a similar pattern between groups, except at the baseline, when insulin was higher in the sleep debt condition of the SR when compared with the SD ( p < 0.01). In the comparison between regular sleep and sleep debt, the SD had a higher insulin AUC ( p < 0.01) and FFAs ( p = 0.03) after sleep deprivation, and insulin and the insulin AUC increased ( p < 0.01 for both), while the ISI decreased ( p = 0.02) after sleep restriction in the SR. In baseline parameters covariate by the condition of regular sleep, insulin ( p = 0.02) and the HOMA-IR ( p < 0.01) were higher, and cortisol ( p = 0.04) was lower after sleep restriction when compared with sleep deprivation. Conclusion Sleep restriction for 4 consecutive nights is more detrimental to energy metabolism because of the higher insulin values and insulin resistance compared with an acute period of sleep deprivation of 24 hours.
Collapse
Affiliation(s)
- Jorge Fernando Tavares Souza
- Departamento de Psicobiologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Marcos Monico-Neto
- Departamento de Psicobiologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Hanna Karen Moreira Antunes
- Departamento de Psicobiologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
- Department of Biosciences, Instituto de Saúde e Sociedade (ISS), Universidade Federal de São Paulo (Unifesp), Santos, SP, Brazil
| |
Collapse
|
9
|
Maloney A, Kanaley JA. Short Sleep Duration Disrupts Glucose Metabolism: Can Exercise Turn Back the Clock? Exerc Sport Sci Rev 2024; 52:77-86. [PMID: 38608214 PMCID: PMC11168896 DOI: 10.1249/jes.0000000000000339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Short sleep duration is prevalent in modern society and may be contributing to type 2 diabetes prevalence. This review will explore the effects of sleep restriction on glycemic control, the mechanisms causing insulin resistance, and whether exercise can offset changes in glycemic control. Chronic sleep restriction may also contribute to a decrease in physical activity leading to further health complications.
Collapse
Affiliation(s)
- Alan Maloney
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | | |
Collapse
|
10
|
Christ JP, Shinkai K, Corley J, Pasch L, Cedars MI, Huddleston HG. Metabolic and endocrine status associate with obstructive sleep apnea risk among patients with polycystic ovary syndrome. J Clin Sleep Med 2024; 20:871-877. [PMID: 38217476 PMCID: PMC11145041 DOI: 10.5664/jcsm.11012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
STUDY OBJECTIVES Risk of obstructive sleep apnea (OSA) appears to be increased among patients with polycystic ovary syndrome (PCOS), but the underlying physiology is unclear. We sought to identify predictors of OSA risk among patients with PCOS. METHODS A cross-sectional analysis of patients evaluated for PCOS at a single tertiary center from 2017-2022 was completed. Inclusion criteria included patients 18-44 years of age who had Rotterdam criteria for PCOS and had completed a Berlin Questionnaire (BQ) for OSA risk assessment. All patients underwent standardized anthropometric, ultrasound, endocrine, and metabolic phenotyping. RESULTS Of the 572 patients screened during the study period, 309 patients with PCOS met inclusion criteria, and 104 (33.7%) had a high-risk BQ. Those with a high-risk BQ, compared with those without, had significantly (P < .05) higher waist:hip ratio, low-density-lipoprotein cholesterol, triglycerides, fasting insulin, 2-hour insulin, fasting glucose, 2-hour glucose, homeostatic model assessment for insulin resistance, hemoglobin A1C, C-reactive protein, free testosterone, and free androgen index and had lower high-density-lipoprotein cholesterol and sex hormone binding globulin. In multivariable modeling controlling for all significantly differing variables in univariate analyses, hemoglobin A1C (β [standard error] 1.05 [0.45], P = .02), C-reactive protein (0.09 [0.04], P = .01), and sex hormone binding globulin (-0.02 [0.01], P = .02) associated with high-risk BQ. CONCLUSIONS Dysglycemia, inflammation, and androgen status independently associate with predicted OSA risk by BQ. Future studies are needed to comprehensively assess the impact of treatment of OSA on these outcomes among patients with PCOS to better clarify the directionality and clinical implications of these associations. CITATION Christ JP, Shinkai K, Corley J, Pasch L, Cedars MI, Huddleston HG. Metabolic and endocrine status associate with obstructive sleep apnea risk among patients with polycystic ovary syndrome. J Clin Sleep Med. 2024;20(6):871-877.
Collapse
Affiliation(s)
- Jacob P. Christ
- Center for Reproductive Health, University of California San Francisco, San Francisco, California
| | - Kanade Shinkai
- Department of Dermatology, University of California San Francisco, San Francisco, California
| | - Jamie Corley
- Center for Reproductive Health, University of California San Francisco, San Francisco, California
| | - Lauri Pasch
- Center for Reproductive Health, University of California San Francisco, San Francisco, California
| | - Marcelle I. Cedars
- Center for Reproductive Health, University of California San Francisco, San Francisco, California
| | - Heather G. Huddleston
- Center for Reproductive Health, University of California San Francisco, San Francisco, California
| |
Collapse
|
11
|
Speksnijder EM, Bisschop PH, Siegelaar SE, Stenvers DJ, Kalsbeek A. Circadian desynchrony and glucose metabolism. J Pineal Res 2024; 76:e12956. [PMID: 38695262 DOI: 10.1111/jpi.12956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Esther M Speksnijder
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Rendine M, Cocci P, de Vivo L, Bellesi M, Palermo FA. Effects of Chronic Sleep Restriction on Transcriptional Sirtuin 1 Signaling Regulation in Male Mice White Adipose Tissue. Curr Issues Mol Biol 2024; 46:2144-2154. [PMID: 38534754 DOI: 10.3390/cimb46030138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic sleep restriction (CSR) is a prevalent issue in modern society that is associated with several pathological states, ranging from neuropsychiatric to metabolic diseases. Despite its known impact on metabolism, the specific effects of CSR on the molecular mechanisms involved in maintaining metabolic homeostasis at the level of white adipose tissue (WAT) remain poorly understood. Therefore, this study aimed to investigate the influence of CSR on sirtuin 1 (SIRT1) and the peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway in the WAT of young male mice. Both genes interact with specific targets involved in multiple metabolic processes, including adipocyte differentiation, browning, and lipid metabolism. The quantitative PCR (qPCR) results demonstrated a significant upregulation of SIRT-1 and some of its target genes associated with the transcriptional regulation of lipid homeostasis (i.e., PPARα, PPARγ, PGC-1α, and SREBF) and adipose tissue development (i.e., leptin, adiponectin) in CSR mice. On the contrary, DNA-binding transcription factors (i.e., CEBP-β and C-myc), which play a pivotal function during the adipogenesis process, were found to be down-regulated. Our results also suggest that the induction of SIRT1-dependent molecular pathways prevents weight gain. Overall, these findings offer new, valuable insights into the molecular adaptations of WAT to CSR, in order to support increased energy demand due to sleep loss.
Collapse
Affiliation(s)
- Marco Rendine
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133 Milan, Italy
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Luisa de Vivo
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy
| | - Michele Bellesi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1QU, UK
| | | |
Collapse
|
13
|
Yu H, Zhao X, Zhang X, Wei H, Zuo A, Guo Y. Sleeping More Hours Per Day Than Working Can Prevent New-Onset Diabetes. Int J Public Health 2023; 68:1606634. [PMID: 38144394 PMCID: PMC10739384 DOI: 10.3389/ijph.2023.1606634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/19/2023] [Indexed: 12/26/2023] Open
Abstract
Objectives: We expressed the combined effect by the ratio of daily sleep time to daily work time. The aim of this study was to discussed the predictive ability of daily sleep hours/work hours (SH/WH) ratio for diabetes risk. Methods: Cox proportional hazards regression was used to calculate the hazard ratios (HRs) of new-onset diabetes. Restricted cubic spline analyses were performed to visualize the influence trend of SH/WH ratio and diabetes risk. Results: The RCS model revealed a non-linear and L-shaped correlation between SH/WH ratio and diabetes risk. Compared with the participates with SH/WH ratio <1, those with a ratio ≥1 had a lower risk of developing diabetes. The multivariable adjusted hazard ratios (HRs) with 95% confidence intervals (CIs) of new-onset diabetes in Q2, Q3, Q4 and Q5 groups compared with Q1 group were 0.82 (0.57, 1.19), 1.05 (0.69, 1.59), 0.57 (0.36, 0.91), 0.66 (0.42, 1.06). The Kaplan-Meier curve showed that Q4 group had lower cumulative incidence. Conclusion: Sleeping longer than working (SH/WH ratio ≥1) can reduce risk for developing diabetes. A minimal risk observed at 1.10-<1.37 (the fourth quintile) of SH/WH ratio.
Collapse
Affiliation(s)
- Haiyan Yu
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoyu Zhao
- Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaodong Zhang
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Haishan Wei
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Anju Zuo
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuan Guo
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
14
|
Azuara-Alvarez LE, Díaz-Muñoz M, Báez Ruiz A, Saderi N, Ramírez-Plascencia OD, Cárdenas-Romero S, Flores-Sandoval O, Salgado-Delgado R. Visceral fat sympathectomy ameliorates systemic and local stress response related to chronic sleep restriction. Exp Biol Med (Maywood) 2023; 248:2381-2392. [PMID: 38143435 PMCID: PMC10903249 DOI: 10.1177/15353702231214267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/23/2023] [Indexed: 12/26/2023] Open
Abstract
Disturbance of sleep homeostasis encompasses health issues, including metabolic disorders like obesity, diabetes, and augmented stress vulnerability. Sleep and stress interact bidirectionally to influence the central nervous system and metabolism. Murine models demonstrate that decreased sleep time is associated with an increased systemic stress response, characterized by endocrinal imbalance, including the elevated activity of hypothalamic-pituitary-adrenal axis, augmented insulin, and reduced adiponectin, affecting peripheral organs physiology, mainly the white adipose tissue (WAT). Within peripheral organs, a local stress response can also be activated by promoting the formation of corticosterone. This local amplifying glucocorticoid signaling is favored through the activation of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). In WAT, 11β-HSD1 activity is upregulated by the sympathetic nervous system, suggesting a link between sleep loss, augmented stress response, and a potential WAT metabolic disturbance. To gain more understanding about this relationship, metabolic and stress responses of WAT-sympathectomized rats were analyzed to identify the contribution of the autonomic nervous system to stress response-related metabolic disorders during chronic sleep restriction. Male Wistar rats under sleep restriction were allowed just 6 h of daily sleep over eight weeks. Results showed that rats under sleep restriction presented higher serum corticosterone, increased adipose tissue 11β-HSD1 activity, weight loss, decreased visceral fat, augmented adiponectin, lower leptin levels, glucose tolerance impairment, and mildly decreased daily body temperature. In contrast, sympathectomized rats under sleep restriction exhibited decreased stress response (lower serum corticosterone and 11β-HSD1 activity). In addition, they maintained weight loss, explained by a reduced visceral fat pad, leptin, and adiponectin, improved glucose management, and persisting decline in body temperature. These results suggest autonomic nervous system is partially responsible for the WAT-exacerbated stress response and its metabolic and physiological disturbances.
Collapse
Affiliation(s)
- Lucia E Azuara-Alvarez
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Querétaro 76230, México
| | - Adrián Báez Ruiz
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Nadia Saderi
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Oscar Daniel Ramírez-Plascencia
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
- Neurology department, Beth Israel Deacones Medical Center/Harvard Medical School, Boston, MA 02215, USA
| | - Skarleth Cárdenas-Romero
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
- Neurology department, Beth Israel Deacones Medical Center/Harvard Medical School, Boston, MA 02215, USA
| | - Omar Flores-Sandoval
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Roberto Salgado-Delgado
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| |
Collapse
|
15
|
Chen W, Xu Y, Li ZH, Si YC, Wang HY, Bian XL, Li L, Guo ZY, Lai XL. Serum metabolic alterations in peritoneal dialysis patients with excessive daytime sleepiness. Ren Fail 2023; 45:2190815. [PMID: 37051665 PMCID: PMC10116928 DOI: 10.1080/0886022x.2023.2190815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Excessive daytime sleepiness (EDS) is associated with quality of life and all-cause mortality in the end-stage renal disease population. This study aims to identify biomarkers and reveal the underlying mechanisms of EDS in peritoneal dialysis (PD) patients. A total of 48 nondiabetic continuous ambulatory peritoneal dialysis patients were assigned to the EDS group and the non-EDS group according to the Epworth Sleepiness Scale (ESS). Ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-Q-TOF/MS) was used to identify the differential metabolites. Twenty-seven (male/female, 15/12; age, 60.1 ± 16.2 years) PD patients with ESS ≥ 10 were assigned to the EDS group, while twenty-one (male/female, 13/8; age, 57.9 ± 10.1 years) PD patients with ESS < 10 were defined as the non-EDS group. With UHPLC-Q-TOF/MS, 39 metabolites with significant differences between the two groups were found, 9 of which had good correlations with disease severity and were further classified into amino acid, lipid and organic acid metabolism. A total of 103 overlapping target proteins of the differential metabolites and EDS were found. Then, the EDS-metabolite-target network and the protein-protein interaction network were constructed. The metabolomics approach integrated with network pharmacology provides new insights into the early diagnosis and mechanisms of EDS in PD patients.
Collapse
Affiliation(s)
- Wei Chen
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Ying Xu
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Zheng-Hao Li
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, P.R. China
| | - Ya-Chen Si
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Hai-Yan Wang
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Xiao-Lu Bian
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Lu Li
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Zhi-Yong Guo
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Xue-Li Lai
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| |
Collapse
|
16
|
Yang K, Hou R, Zhao J, Wang X, Wei J, Pan X, Zhu X. Lifestyle effects on aging and CVD: A spotlight on the nutrient-sensing network. Ageing Res Rev 2023; 92:102121. [PMID: 37944707 DOI: 10.1016/j.arr.2023.102121] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/12/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
Aging is widespread worldwide and a significant risk factor for cardiovascular disease (CVD). Mechanisms underlying aging have attracted considerable attention in recent years. Remarkably, aging and CVD overlap in numerous ways, with deregulated nutrient sensing as a common mechanism and lifestyle as a communal modifier. Interestingly, lifestyle triggers or suppresses multiple nutrient-related signaling pathways. In this review, we first present the composition of the nutrient-sensing network (NSN) and its metabolic impact on aging and CVD. Secondly, we review how risk factors closely associated with CVD, including adverse life states such as sedentary behavior, sleep disorders, high-fat diet, and psychosocial stress, contribute to aging and CVD, with a focus on the bridging role of the NSN. Finally, we focus on the positive effects of beneficial dietary interventions, specifically dietary restriction and the Mediterranean diet, on the regulation of nutrient metabolism and the delayed effects of aging and CVD that depend on the balance of the NSN. In summary, we expound on the interaction between lifestyle, NSN, aging, and CVD.
Collapse
Affiliation(s)
- Kaiying Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Rongyao Hou
- Department of Neurology, The Affiliated Hiser Hospital of Qingdao University, Qingdao 266000, China
| | - Jie Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xia Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
17
|
Sengupta A, Tudor JC, Cusmano D, Baur JA, Abel T, Weljie AM. Sleep deprivation and aging are metabolically linked across tissues. Sleep 2023; 46:zsad246. [PMID: 37738102 PMCID: PMC11502955 DOI: 10.1093/sleep/zsad246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/21/2023] [Indexed: 09/24/2023] Open
Abstract
STUDY OBJECTIVES Insufficient sleep is a concerning hallmark of modern society because sleep deprivation (SD) is a risk factor for neurodegenerative and cardiometabolic disorders. SD imparts an aging-like effect on learning and memory, although little is known about possible common molecular underpinnings of SD and aging. Here, we examine this question by profiling metabolic features across different tissues after acute SD in young adult and aged mice. METHODS Young adult and aged mice were subjected to acute SD for 5 hours. Blood plasma, hippocampus, and liver samples were subjected to UPLC-MS/MS-based metabolic profiling. RESULTS SD preferentially impacts peripheral plasma and liver profiles (e.g. ketone body metabolism) whereas the hippocampus is more impacted by aging. We further demonstrate that aged animals exhibit SD-like metabolic features at baseline. Hepatic alterations include parallel changes in nicotinamide metabolism between aging and SD in young animals. Overall, metabolism in young adult animals is more impacted by SD, which in turn induces aging-like features. A set of nine metabolites was classified (79% correct) based on age and sleep status across all four groups. CONCLUSIONS Our metabolic observations demonstrate striking parallels to previous observations in studies of learning and memory and define a molecular metabolic signature of sleep loss and aging.
Collapse
Affiliation(s)
- Arjun Sengupta
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current affiliation: Department of Biology, Saint Joseph’s University, Philadelphia, PA, USA
| | - Danielle Cusmano
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
- Current Affiliation: Iowa Neuroscience Institute, Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, 2312 PBDB, Iowa City, IA, USA
| | - Aalim M Weljie
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Barragán R, Zuraikat FM, Cheng B, Scaccia SE, Cochran J, Aggarwal B, Jelic S, St‐Onge M. Paradoxical Effects of Prolonged Insufficient Sleep on Lipid Profile: A Pooled Analysis of 2 Randomized Trials. J Am Heart Assoc 2023; 12:e032078. [PMID: 37815115 PMCID: PMC10757551 DOI: 10.1161/jaha.123.032078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 10/11/2023]
Abstract
Background Insufficient sleep is associated with increased cardiovascular disease risk, but causality is unclear. We investigated the impact of prolonged mild sleep restriction (SR) on lipid and inflammatory profiles. Methods and Results Seventy-eight participants (56 women [12 postmenopausal]; age, 34.3±12.5 years; body mass index, 25.8±3.5 kg/m2) with habitual sleep duration 7 to 9 h/night (adequate sleep [AS]) underwent two 6-week conditions in a randomized crossover design: AS versus SR (AS-1.5 h/night). Total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol, triglycerides, and inflammatory markers (CRP [C-reactive protein], interleukin 6, and tumor necrosis factor-α) were assessed. Linear models tested effects of SR on outcomes in the full sample and by sex+menopausal status (premenopausal versus postmenopausal women+men). In the full sample, SR increased high-density lipoprotein cholesterol compared with AS (β=1.2±0.5 mg/dL; P=0.03). Sex+menopausal status influenced the effects of SR on change in total cholesterol (P-interaction=0.04), LDL-C (P-interaction=0.03), and interleukin 6 (P-interaction=0.07). Total cholesterol and LDL-C decreased in SR versus AS in premenopausal women (total cholesterol: β=-4.2±1.9 mg/dL; P=0.03; LDL-C: β=-6.3±2.0 mg/dL; P=0.002). Given paradoxical effects of SR on cholesterol concentrations, we explored associations between changes in inflammation and end point lipids under each condition. Increases in interleukin 6 and tumor necrosis factor-α during SR tended to relate to lower LDL-C in premenopausal women (interleukin 6: β=-5.3±2.6 mg/dL; P=0.051; tumor necrosis factor-α: β=-32.8±14.2 mg/dL; P=0.027). Conclusions Among healthy adults, prolonged insufficient sleep does not increase atherogenic lipids. However, increased inflammation in SR tends to predict lower LDL-C in premenopausal women, resembling the "lipid paradox" in which low cholesterol associates with increased cardiovascular disease risk in proinflammatory conditions. Registration URL: https://www.clinicaltrials.gov; Unique identifiers: NCT02835261, NCT02960776.
Collapse
Affiliation(s)
- Rocío Barragán
- Department of Preventive Medicine and Public HealthUniversity of ValenciaValenciaSpain
- Centro de Investigación Biomédica En Red Fisiopatología de la Obesidad y NutriciónInstituto de Salud Carlos IIIMadridSpain
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
| | - Faris M. Zuraikat
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of General Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
- New York Nutrition Obesity Research CenterColumbia University Irving Medical CenterNew YorkNY
| | - Bin Cheng
- Department of Biostatistics, Mailman School of Public HealthColumbia University Irving Medical CenterNew YorkNY
| | - Samantha E. Scaccia
- Division of Cardiology, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Justin Cochran
- Department of SurgeryColumbia University Irving Medical CenterNew YorkNY
| | - Brooke Aggarwal
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of Cardiology, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Sanja Jelic
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Marie‐Pierre St‐Onge
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of General Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
- New York Nutrition Obesity Research CenterColumbia University Irving Medical CenterNew YorkNY
| |
Collapse
|
19
|
Tajiri E, Yoshimura E, Tobina T, Yamashita T, Kume K, Hatamoto Y, Shimoda S. Effects of sleep restriction on food intake and appetite under free-living conditions: A randomized crossover trial. Appetite 2023; 189:106998. [PMID: 37562755 DOI: 10.1016/j.appet.2023.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
To investigate changes in subjective psychological factors and dietary intake during sleep restriction, we carried out a randomized crossover trial with a 3-day sleep restriction condition (SR; 5 h of sleep) and control sleep condition (CS; 8 h of sleep). Days 3 and 4 involved free-living and laboratory (in the morning) conditions, respectively. Subjective psychological factors (hunger, appetite, desire for sweets and fatty foods, sleepiness, and fatigue) were assessed using a 0.0-10.0 cm visual analog scale (VAS) every hour throughout the day on day 3, and at 8:00 a.m. on day 4. Dietary intake on day 3 was assessed on the basis of the food purchased and eaten. Fasting blood samples were collected at 8:00 a.m. on day 4. Dietary intake during the ad libitum breakfast was assessed on day 4. The participants were 13 women and 11 men (mean age, 21.4 ± 1.0 years; mean body mass index, 19.8 ± 1.7 kg/m2). The areas under the curve 0-16 h after waking for hunger, desire for fatty foods, sleepiness, and fatigue were higher in the SR than CS on day 3 (P < 0.05). Energy and carbohydrate intakes from snacks (daytime and nighttime) on day 3 were higher in the SR than CS (P < 0.05) but total dietary intake on day 3 was not different between the conditions (P > 0.05). The 2-arachidonoylglycerol level was different between the conditions (P < 0.05), but was not associated with sweet taste preference, dietary intake, or the active ghrelin level on day 4 (P > 0.05). In conclusion, ratings for subjective psychological factors and energy and carbohydrate intakes from snacks increased in association with sleep restriction under free-living conditions.
Collapse
Affiliation(s)
- Eri Tajiri
- Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, 862-8502, Japan.
| | - Eiichi Yoshimura
- Department of Nutrition and Metabolism, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 566-0002, Japan.
| | - Takuro Tobina
- Faculty of Nursing and Nutrition, University of Nagasaki, Nagasaki, 851-2195, Japan.
| | - Tomoki Yamashita
- Faculty of Nursing and Nutrition, University of Nagasaki, Nagasaki, 851-2195, Japan.
| | - Kokoro Kume
- Faculty of Nursing and Nutrition, University of Nagasaki, Nagasaki, 851-2195, Japan.
| | - Yoichi Hatamoto
- Department of Nutrition and Metabolism, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 566-0002, Japan.
| | - Seiya Shimoda
- Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, Kumamoto, 862-8502, Japan.
| |
Collapse
|
20
|
Singh P, Beyl RA, Stephens JM, Noland RC, Richard AJ, Boudreau A, Hebert RC, Ravussin E, Broussard JL, St-Onge MP, Marlatt KL. Effect of sleep restriction on insulin sensitivity and energy metabolism in postmenopausal women: A randomized crossover trial. Obesity (Silver Spring) 2023; 31:1204-1215. [PMID: 36998155 PMCID: PMC10191900 DOI: 10.1002/oby.23739] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/10/2023] [Accepted: 01/26/2023] [Indexed: 04/01/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the effect of sleep restriction (SR) on insulin sensitivity and energy metabolism in postmenopausal women. METHODS In a randomized crossover trial, 14 women underwent four nights of habitual sleep (HS, 100% normal sleep) and SR (60% of HS) while following a eucaloric diet. Outcomes included the following: (1) insulin sensitivity by hyperinsulinemic-euglycemic clamp, defined as the glucose infusion rate (GIR); (2) resting metabolism and substrate oxidation by indirect calorimetry; and (3) glucose, insulin, and C-peptide concentrations following a standard meal test. RESULTS Nine postmenopausal women (mean [SD], age 59 [4] years, BMI 28.0 [2.6] kg/m2 ) were analyzed. Accelerometer-determined total time in bed was 8.4 ± 0.6 hours during HS versus 5.0 ± 0.4 hours during SR (38% reduction, p < 0.0001). SR reduced low-dose insulin GIR by 20% (HS: 2.55 ± 0.22 vs. SR: 2.03 ± 0.20 mg/kg/min; p = 0.01) and high-dose insulin GIR by 12% (HS: 10.48 ± 0.72 vs. SR: 9.19 ± 0.72 mg/kg/min; p < 0.001). SR reduced fat oxidation during high-dose insulin infusion (p < 0.01), and it did not alter resting energy metabolism. CONCLUSIONS Four nights of SR reduced insulin sensitivity and fat oxidation in postmenopausal women. These findings underscore the role of insufficient sleep in metabolic dysfunction following menopause. Larger trials investigating how sleep disturbances cause metabolic dysfunction during menopause are needed across all stages of menopause.
Collapse
Affiliation(s)
- Prachi Singh
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Robbie A. Beyl
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | | | | - Anik Boudreau
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Josiane L. Broussard
- Department of Health & Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Marie-Pierre St-Onge
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
21
|
El-Taravi Y, Baimukhambetova DV, Gorlenko CL, Kiseljow HY, Kokhanovskaia II, Burotina AI, Rassadina ES, Martirosian NS, Petunina NA. Management of diabetes during Ramadan: an update for Russian-speaking doctors. DIABETES MELLITUS 2023. [DOI: 10.14341/dm12972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Fasting during the Islamic Ramadan month is one of the five obligatory pillars for each adult, healthy, and sane Muslim. People with severe illnesses, including type 1 and type 2 diabetes mellitus are exempt from fasting. However, many Muslims often insist on Ramadan participating despite any medical advises. It’s known that Muslims are the second largest religious group in Russia; thus, its crucial to have as much modern recommendation for management patients with type 1 and type 2 diabetes mellitus as possible. The aim of this narrative review is to evaluate physiological and pathophysiological metabolism changing during holy Ramadan month, to clarify the management of patients with type 1 and type 2 diabetes mellitus during Ramadan, to determine the frequency of glucose measuring during the day, and to understand when its time to interrupt the fasting. Moreover, we discuss specific recommendations in glucose-lowering therapy changing, nutrition, physical activities and education.
Collapse
Affiliation(s)
- Y.A. El-Taravi
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | | - C. L. Gorlenko
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - H. Yu. Kiseljow
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | | - A. I. Burotina
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. S. Rassadina
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - N. S. Martirosian
- I.M. Sechenov First Moscow State Medical University (Sechenov University); L.A. Vorokhobov City Clinical Hospital No. 67
| | - N. A. Petunina
- I.M. Sechenov First Moscow State Medical University (Sechenov University); L.A. Vorokhobov City Clinical Hospital No. 67
| |
Collapse
|
22
|
Duan D, Kim LJ, Jun JC, Polotsky VY. Connecting insufficient sleep and insomnia with metabolic dysfunction. Ann N Y Acad Sci 2023; 1519:94-117. [PMID: 36373239 PMCID: PMC9839511 DOI: 10.1111/nyas.14926] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The global epidemic of obesity and type 2 diabetes parallels the rampant state of sleep deprivation in our society. Epidemiological studies consistently show an association between insufficient sleep and metabolic dysfunction. Mechanistically, sleep and circadian rhythm exert considerable influences on hormones involved in appetite regulation and energy metabolism. As such, data from experimental sleep deprivation in humans demonstrate that insufficient sleep induces a positive energy balance with resultant weight gain, due to increased energy intake that far exceeds the additional energy expenditure of nocturnal wakefulness, and adversely impacts glucose metabolism. Conversely, animal models have found that sleep loss-induced energy expenditure exceeds caloric intake resulting in net weight loss. However, animal models have significant limitations, which may diminish the clinical relevance of their metabolic findings. Clinically, insomnia disorder and insomnia symptoms are associated with adverse glucose outcomes, though it remains challenging to isolate the effects of insomnia on metabolic outcomes independent of comorbidities and insufficient sleep durations. Furthermore, both pharmacological and behavioral interventions for insomnia may have direct metabolic effects. The goal of this review is to establish an updated framework for the causal links between insufficient sleep and insomnia and risks for type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Daisy Duan
- Division of Endocrinology, Diabetes, and Metabolism; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lenise J. Kim
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan C. Jun
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care; Department of Medicine; Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
23
|
Brager AJ, Gordon JA, Rouska A, Moore B, Mysliwiec V. Circadian Advantages in Elite Athletes. CURRENT SLEEP MEDICINE REPORTS 2022. [DOI: 10.1007/s40675-022-00239-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
24
|
Sivakumaran K, Ritonja JA, Palmer N, Pasumarthi T, Waseem H, Yu T, Denning A, Michaud D, Morgan RL. Effect of sleep disturbance on biomarkers related to the development of adverse health outcomes: A systematic review of the human literature. J Sleep Res 2022; 32:e13775. [PMID: 36330773 DOI: 10.1111/jsr.13775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/11/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
Abstract
Literature suggests that unrestricted and undisturbed sleep is vital for basic human function and performance; however, it is unclear as to what amount of sleep disturbance leads to dysregulation in biomarkers, which may underscore the development of adverse health effects. This systematic review aims to identify the amount of sleep disturbance that contributes to biomarker changes as a potential precursor to the development of adverse health effects. English-language comparative studies available in PubMed, Cochrane Central, EMBASE, and CINAHL databases from 1 January 1980 to 31 July 2021 were searched. Where possible, random-effects meta-analyses were used to examine the effect of sleep disturbances on adverse health effects. The risk of bias of individual studies was assessed using the Cochrane Risk of Bias Tool and the Risk of Bias of Nonrandomised Studies - of Exposures instruments and the certainty of the body of evidence for each outcome was assessed using the Grading of Recommendations Assessment, Development and Evaluation approach. The search identified 92 primary studies reporting on blood pressure, hypertension, heart rate, cardiac arrhythmia, cardiac output, waist circumference, cortisol, adrenaline, noradrenaline, immune system markers, glucose, insulin, cholesterol, and triglyceride levels. Although some meta-analyses suggested there may be an association between sleep disturbances and certain outcomes, the certainty in the evidence was very low due to concerns with risk of bias, inconsistency across exposures, populations, and imprecision in the estimates of effects. Further research is needed to explore the point at which types, levels and duration of sleep disturbances may begin to increase the risk of developing adverse health outcomes to inform and tailor health interventions.
Collapse
Affiliation(s)
| | - Jennifer A. Ritonja
- Université de Montréal Hospital Research Centre (CRCHUM) Montreal Quebec Canada
- Department of Social and Preventive Medicine Université de Montréal Montreal Quebec Canada
| | | | - Tejanth Pasumarthi
- Evidence Foundation Cleveland Heights Ohio USA
- School of Interdisciplinary Science McMaster University Hamilton Ontario Canada
| | - Haya Waseem
- Evidence Foundation Cleveland Heights Ohio USA
| | - Tiffany Yu
- Evidence Foundation Cleveland Heights Ohio USA
- Faculty of Health Sciences McMaster University Hamilton Ontario Canada
| | - Allison Denning
- Health Canada, Environmental and Radiation Health Sciences Directorate Consumer & Clinical Radiation Protection Bureau Ottawa Ontario Canada
| | - David Michaud
- Health Canada, Environmental and Radiation Health Sciences Directorate Consumer & Clinical Radiation Protection Bureau Ottawa Ontario Canada
| | - Rebecca L. Morgan
- Evidence Foundation Cleveland Heights Ohio USA
- Department of Health Research Methods, Evidence and Impact McMaster University Hamilton Ontario Canada
| |
Collapse
|
25
|
Wang C, Huang T, Song W, Zhu J, Liu Y, Chen X, Sun X, Wu Q, Chen H, Liao H, Lin J, Ou X, Zou Z, Wang Z, Zheng Z, Wu K, Chen R. A meta-analysis of the relationship between polycystic ovary syndrome and sleep disturbances risk. Front Physiol 2022; 13:957112. [PMID: 36246113 PMCID: PMC9558285 DOI: 10.3389/fphys.2022.957112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
Objective: A meta-analysis is used to explore the relationship between polycystic ovary syndrome (PCOS) and the risk of Sleep disturbances. Method: Cochrane Library, PubMed, Embase, and Web of Science databases are searched by computer from their establishment to 1 May 2022. Review Manager 5.4 software is used for the meta-analysis. Results: A total of nine articles are included, with 1,107 subjects. The results show that PCOS is positively associated with the risk of Sleep disturbances. Comparing with the “PCOS group” (experimental group) with the “NON-PCOS group” (control group), the incidence of Sleep disturbances is higher (OR = 11.24, 95% CI: 2.00–63.10, Z = 2.75, p = 0.006); the Pittsburgh Sleep Quality Index (PSQI) scores of the PCOS group is higher than that of the NON-PCOS group (MD = 0.78, 95% CI: 0.32–1.25, Z = 3.30, p = 0.001); the Epworth Sleepiness Scale (ESS) scores of the PCOS group is higher than that of the NON-PCOS group (MD = 2.49, 95% CI: 0.80–4.18, Z = 2.88, p = 0.004); Apnea hypopnea index (AHIs) in the PCOS group are higher than those in the NON-PCOS group (MD = 2.68, 95% CI: 1.07–4.28, Z = 3.27, p = 0.001); the sleep efficiency of the PCOS group is lower than that of the NON-PCOS group (MD = -5.16, 95% CI: 9.39–-0.93, Z = 2.39, p = 0.02); the sleep onset latency of the PCOS group is higher than that of the NON-PCOS group (MD = 2.45, 95% CI: 1.40–3.50, Z = 4.57, p < 0.001); and the Rapid Eyes Movement (REM) sleep in the PCOS group is higher than that in the NON-PCOS group (MD = 17.19, 95% CI: 11.62–55.76, Z = 6.05, p < 0.001). The studies included in each analysis have publication biases of different sizes. After subgroup analysis and sensitivity analysis, the heterogeneity of each study in the meta-analysis is reduced, the bias is reduced accordingly, and the stability of the results can be maintained. Conclusion: PCOS is positively associated with the risk of Sleep disturbances. In order to reduce such risk, attention should be paid to the role of PCOS management, and PCOS prevention and treatment should be actively carried out.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Respiratory and Critical Care Medicine, Taishan Hospital of Traditional Chinese Medicine, Jiangmen, China
| | - Tao Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wu Song
- Department of Laboratory, Dongguan Maternal and Child Health Care Hospital, Dongguan, China
| | - Jinru Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yanhong Liu
- Department of Gynecology, Taishan Hospital of Traditional Chinese Medicine, Jiangmen, China
| | | | - Xishi Sun
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qinglan Wu
- Department of Respiratory and Critical Care Medicine, Central People’s Hospital of Zhanjiang, Zhanjiang, China
| | - Huimin Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huizhao Liao
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junyan Lin
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xudong Ou
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhihong Zou
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Wang
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenzhen Zheng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- *Correspondence: Zhenzhen Zheng, ; Kang Wu, ; Riken Chen,
| | - Kang Wu
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Zhenzhen Zheng, ; Kang Wu, ; Riken Chen,
| | - Riken Chen
- Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Zhenzhen Zheng, ; Kang Wu, ; Riken Chen,
| |
Collapse
|
26
|
Teo P, Henry BA, Moran LJ, Cowan S, Bennett C. The role of sleep in PCOS: what we know and what to consider in the future. Expert Rev Endocrinol Metab 2022; 17:305-318. [PMID: 35815469 DOI: 10.1080/17446651.2022.2082941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Sleep disturbance and clinical sleep conditions disrupt endocrine signals, energy expenditure and nutritional intake. Women with polycystic ovary syndrome (PCOS) are at higher risk of sleep disturbances and clinical conditions. It is possible that sleep may contribute to the exacerbation of PCOS. This review aims to explore the relationship between sleep and chronic disease, particularly in women with PCOS. AREAS COVERED This review narratively explores what sleep is, how to measure sleep and the possible mechanisms that support the link between sleep in adipose tissue deposition, insulin resistance and the presentation of PCOS. EXPERT OPINION Research shows that disturbed sleep and clinical sleep conditions disrupt energy expenditure. This may increase adipose tissue deposition and exacerbate insulin resistance which are known to worsen the presentation of PCOS. Further, sleep disturbance in women with PCOS may ameliorate any positive lifestyle changes made after diagnosis. Cognitive behavioural therapy interventions for sleep are a successful strategy for the management of sleep disturbances in the general population. However, such interventions are yet to be trialled in women with PCOS. Given the proposed implications, interventions to improve sleep could provide additional support for women with PCOS to successfully implement lifestyle strategies and should be further investigated.
Collapse
Affiliation(s)
- Peiseah Teo
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Belinda A Henry
- Department of Physiology, Monash University, Melbourne, VIC, Australia
- Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Lisa J Moran
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | | | - Christie Bennett
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Kanagasabai T, Riddell MC, Ardern CI. Inflammation, Oxidative Stress, and Antioxidant Micronutrients as Mediators of the Relationship Between Sleep, Insulin Sensitivity, and Glycosylated Hemoglobin. Front Public Health 2022; 10:888331. [PMID: 35757614 PMCID: PMC9218604 DOI: 10.3389/fpubh.2022.888331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Sleep deprivation and poor sleep quality contribute to increases in oxidative stress, antioxidant imbalance, and a pro-inflammatory state which may predispose to a higher risk of diabetes. Our objective was to estimate the contributions of C-reactive protein (CRP), gamma glutamyl transferase (GGT), and micronutrient antioxidants (bilirubin, carotenoids, uric acid, vitamins A, C-E?) to the relationships between sleep-fasting insulin concentration and -glycosylated hemoglobin (HbA1c). Methods Data from the 2005/06 US National Health and Nutritional Examination Survey were used (N = 1,946; 20 y+). Sleep quality and quantity was assessed by the Sleep Disorders Questionnaire, and fasting blood was collected to quantify CRP, GGT, antioxidant micronutrients, insulin concentration, and HbA1c. The bootstrap method was used to estimate the amount of mediation or contribution of these mediators to the sleep-insulin concentration and -HbA1c relationships, which were quantified as large (≥0.25) or moderate (≥0.09). Results The sleep duration-fasting insulin relationship was mediated by GGT, carotenoids, uric acid, and vitamins C and D, whereas CRP and bilirubin were non-significant mediators of a moderate effect size. Similarly, the sleep quality-fasting insulin relationship was mediated by CRP, bilirubin and vitamin C, whereas GGT, carotenoids, uric acid, and vitamin D were non-significant large-to-moderate mediators. To a lesser degree, these micronutrients mediated for the relationship between sleep-HbA1c levels. Conclusion Several factors related to inflammation, oxidative stress, and antioxidant status were found to lie on the pathway of the sleep-insulin and -glycemic control relationships. Sleep hygiene, reduced systemic inflammation/oxidative stress, and optimal antioxidants intake are potentially beneficial targets for managing diabetes risk.
Collapse
Affiliation(s)
| | - Michael C Riddell
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Chris I Ardern
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| |
Collapse
|
28
|
Abstract
Sleep loss has negative impacts on quality of life, mood, cognitive function and heath. Insomnia or difficulty sleeping is also a prevalent issue, affecting up to 35% of the population at some point in their lives. Insomnia is linked to poor mood, increased use of health care resources, and decreased quality of life as well as possible links to cardiovascular risk factors and disease. Studies have shown an increase in cortisol levels, decreased immunity, and increased markers of sympathetic activity in sleep-deprived healthy subjects and those with chronic insomnia. The literature also shows that subjective complaints consistent with chronic insomnia and shortened sleep time, both independently and in combination, can be associated with the development of diabetes, hypertension, and cardiovascular disease. In this article, we will explore the relationship and strength of association between insufficient sleep and insomnia with these health conditions.
Collapse
Affiliation(s)
- Meena S Khan
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA; Department of Neurology, The Ohio State University, Columbus, OH, USA.
| | - Rita Aouad
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
29
|
Habib S, Sangaraju SL, Yepez D, Grandes XA, Talanki Manjunatha R. The Nexus Between Diabetes and Depression: A Narrative Review. Cureus 2022; 14:e25611. [PMID: 35784974 PMCID: PMC9249007 DOI: 10.7759/cureus.25611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 12/30/2022] Open
Abstract
Comorbid diabetes and depression are a significant public health burden as the consequences of both diseases are worsened by each other. In this study, we have compiled and analyzed findings from various studies to demonstrate that diabetes has a strong association with depression. Both have a significant impact on the quality of life, although the exact mechanisms through which these two chronic diseases affect each other remain unknown. This article discussed the shared etiological factors of comorbidity between diabetes and depression, including physiological (e.g., deregulation of the hypothalamic-pituitary-adrenal (HPA) axis, sympathetic nervous system (SNS) overactivity, microvascular dysfunction, arterial stiffening, inflammation, and cytokines), behavioral (e.g., diet and lifestyle modifications), and environmental (e.g., childhood adversity, poverty, and neighborhood environment). Included data from a range of settings have suggested that the prognosis of both diabetes and depression, in terms of complications, treatment efficacy, morbidity, and mortality, is worse for either disease when they occur concurrently than individually. The implication for the physical, mental, and social well-being of depression in diabetes causes poor self-care and adherence to medical treatment. This article also highlights the importance of regular screening and prompts the treatment of comorbid diabetes and depression with pharmacotherapy, face-to-face psychotherapy, and non-face-to-face models of alternative psychological interventions, including information and communication technologies (ICTs), computer-based diabetes self-management interventions, and digital mental health intervention, to improve the outcomes of both diseases.
Collapse
Affiliation(s)
- Salma Habib
- Internal Medicine, Institute of Applied Health Sciences (IAHS), Chittagong, BGD
| | - Sai Lahari Sangaraju
- Research, People's Education Society (PES) Institute of Medical Sciences and Research, Kuppam, IND
| | - Daniela Yepez
- General Medicine, Universidad Catolica de Santiago de Guayaquil, Guayaquil, ECU
| | - Xavier A Grandes
- General Medicine, Universidad Catolica de Santiago de Guayaquil, Guayaquil, ECU
| | | |
Collapse
|
30
|
Abdollahi A, Dowden BN, Buhman KK, Zembroski AS, Henderson GC. Albumin knockout mice exhibit reduced plasma free fatty acid concentration and enhanced insulin sensitivity. Physiol Rep 2022; 10:e15161. [PMID: 35238481 PMCID: PMC8892599 DOI: 10.14814/phy2.15161] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 04/15/2023] Open
Abstract
Circulating albumin is expected to play a significant role in the trafficking of plasma free fatty acids (FFA) between tissues, such as FFA transfer from adipose tissue to the liver. However, it was not yet known how disrupting FFA binding to albumin in circulation would alter lipid metabolism and any resulting impacts upon control of glycemia. To improve understanding of metabolic control, we aimed to determine whether lack of serum albumin would decrease plasma FFA, hepatic lipid storage, whole body substrate oxidation, and glucose metabolism. Male and female homozygous albumin knockout mice and C57BL/6J wild type controls, each on a standard diet containing a moderate fat content, were studied at 6-8 weeks of age. Indirect calorimetry, glucose tolerance testing, insulin tolerance testing, exercise performance, plasma proteome, and tissue analyses were performed. In both sexes of albumin knockout mice compared to the wild type mice, significant reductions (p < 0.05) were observed for plasma FFA concentration, hepatic triacylglycerol and diacylglycerol content, blood glucose during the glucose tolerance test, and blood glucose during the insulin tolerance test. Albumin deficiency did not reduce whole body fat oxidation over a 24-h period and did not alter exercise performance in an incremental treadmill test. The system-level phenotypic changes in lipid and glucose metabolism were accompanied by reduced hepatic perilipin-2 expression (p < 0.05), as well as increased expression of adiponectin (p < 0.05) and glucose transporter-4 (p < 0.05) in adipose tissue. The results indicate an important role of albumin and plasma FFA concentration in lipid metabolism and glucoregulation.
Collapse
Affiliation(s)
- Afsoun Abdollahi
- Department of Nutrition SciencePurdue UniversityWest LafayetteIndianaUSA
| | - Brianna N. Dowden
- Department of Nutrition SciencePurdue UniversityWest LafayetteIndianaUSA
| | - Kimberly K. Buhman
- Department of Nutrition SciencePurdue UniversityWest LafayetteIndianaUSA
| | | | | |
Collapse
|
31
|
Effects of sleep manipulation on markers of insulin sensitivity: a systematic review and meta-analysis of randomized controlled trials. Sleep Med Rev 2022; 62:101594. [DOI: 10.1016/j.smrv.2022.101594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 01/03/2023]
|
32
|
Kass L, Sanderson JC, Desai T, Hurst R. The relationship between the elevation of haemoglobin A1c level, sleep quality and sleep duration in clinically diagnosed pre-diabetic patients in a nationally representative sample. Diab Vasc Dis Res 2022; 19:14791641211067421. [PMID: 35166578 PMCID: PMC8851951 DOI: 10.1177/14791641211067421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND/OBJECTIVES Type 2 diabetes mellitus (T2DM) is one of the most common chronic illnesses in the United Kingdom accounting for approximately 15% of deaths per year. Growing evidence suggests that sleep duration and quality contributes towards this. This study aimed to determine whether there was a significant relationship between the elevation of haemoglobin A1c (HbA1c) level, sleep quality (SQ) and sleep duration (SD) in clinically diagnosed pre-diabetic patients. SUBJECTS/METHODS Following referral from a relevant healthcare professional, participants (n = 40) were registered on the National Health Service England, funded Healthier You: National Diabetes Prevention Programme and completed a Pittsburgh Sleep Quality Index questionnaire to evaluate SQ and SD. RESULTS A Spearman's correlation showed an association between HbA1c, SQ and SD measures. A simple linear regression showed a significant large positive association (rs = 0.913, p < 0.001) and significant regression (F (1) = 39, p < 0.001) with an R2 of 0.842 between HbA1c level and SQ. Additionally, a significant large negative association (rs = 0.757, p < 0.001) and significant regression was found (F (1) = 39, p < 0.001) with an R2 of 0.570 between HbA1c and SD. CONCLUSIONS This study suggests a relationship between SQ, SD and the elevation of HbA1c which may contribute towards prevalence of T2DM and may help to increase adherence to diabetes prevention programmes.
Collapse
Affiliation(s)
- Lindsy Kass
- School of Life and Medical Science, University of Hertfordshire, Hatfield, UK
- Lindsy Kass, School of Life and Medical Science, University of Hertfordshire, Institute of Sport, Hatfield AL10 9EU, UK.
| | - Julia C Sanderson
- School of Life and Medical Science, University of Hertfordshire, Hatfield, UK
| | - Terun Desai
- School of Life and Medical Science, University of Hertfordshire, Hatfield, UK
| | - Rebecca Hurst
- School of Life and Medical Science, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
33
|
Lin W, Saner NJ, Weng X, Caruana NJ, Botella J, Kuang J, Lee MJC, Jamnick NA, Pitchford NW, Garnham A, Bartlett JD, Chen H, Bishop DJ. The Effect of Sleep Restriction, With or Without Exercise, on Skeletal Muscle Transcriptomic Profiles in Healthy Young Males. Front Endocrinol (Lausanne) 2022; 13:863224. [PMID: 35937838 PMCID: PMC9355502 DOI: 10.3389/fendo.2022.863224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Inadequate sleep is associated with many detrimental health effects, including increased risk of developing insulin resistance and type 2 diabetes. These effects have been associated with changes to the skeletal muscle transcriptome, although this has not been characterised in response to a period of sleep restriction. Exercise induces a beneficial transcriptional response within skeletal muscle that may counteract some of the negative effects associated with sleep restriction. We hypothesised that sleep restriction would down-regulate transcriptional pathways associated with glucose metabolism, but that performing exercise would mitigate these effects. METHODS 20 healthy young males were allocated to one of three experimental groups: a Normal Sleep (NS) group (8 h time in bed per night (TIB), for five nights (11 pm - 7 am)), a Sleep Restriction (SR) group (4 h TIB, for five nights (3 am - 7 am)), and a Sleep Restriction and Exercise group (SR+EX) (4 h TIB, for five nights (3 am - 7 am) and three high-intensity interval exercise (HIIE) sessions (performed at 10 am)). RNA sequencing was performed on muscle samples collected pre- and post-intervention. Our data was then compared to skeletal muscle transcriptomic data previously reported following sleep deprivation (24 h without sleep). RESULTS Gene set enrichment analysis (GSEA) indicated there was an increased enrichment of inflammatory and immune response related pathways in the SR group post-intervention. However, in the SR+EX group the direction of enrichment in these same pathways occurred in the opposite directions. Despite this, there were no significant changes at the individual gene level from pre- to post-intervention. A set of genes previously shown to be decreased with sleep deprivation was also decreased in the SR group, but increased in the SR+EX group. CONCLUSION The alterations to inflammatory and immune related pathways in skeletal muscle, following five nights of sleep restriction, provide insight regarding the transcriptional changes that underpin the detrimental effects associated with sleep loss. Performing three sessions of HIIE during sleep restriction attenuated some of these transcriptional changes. Overall, the transcriptional alterations observed with a moderate period of sleep restriction were less evident than previously reported changes following a period of sleep deprivation.
Collapse
Affiliation(s)
- Wentao Lin
- College of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Nicholas J. Saner
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Human Integrative Physiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiquan Weng
- College of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Nikeisha J. Caruana
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Javier Botella
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Jujiao Kuang
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Matthew J-C. Lee
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Nicholas A. Jamnick
- Metabolic Research Unit, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Nathan W. Pitchford
- School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Andrew Garnham
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | | | - Hao Chen
- College of Exercise and Health, Guangzhou Sport University, Guangzhou, China
- *Correspondence: Hao Chen, ; David J. Bishop,
| | - David J. Bishop
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- *Correspondence: Hao Chen, ; David J. Bishop,
| |
Collapse
|
34
|
Antza C, Kostopoulos G, Mostafa S, Nirantharakumar K, Tahrani A. The links between sleep duration, obesity and type 2 diabetes mellitus. J Endocrinol 2021; 252:125-141. [PMID: 34779405 PMCID: PMC8679843 DOI: 10.1530/joe-21-0155] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/12/2021] [Indexed: 11/24/2022]
Abstract
Global rates of obesity and type 2 diabetes mellitus (T2DM) are increasing globally concomitant with a rising prevalence of sleep deprivation and sleep disorders. Understanding the links between sleep, obesity and T2DM might offer an opportunity to develop better prevention and treatment strategies for these epidemics. Experimental studies have shown that sleep restriction is associated with changes in energy homeostasis, insulin resistance and β-cell function. Epidemiological cohort studies established short sleep duration as a risk factor for developing obesity and T2DM. In addition, small studies suggested that short sleep duration was associated with less weight loss following lifestyle interventions or bariatric surgery. In this article, we review the epidemiological evidence linking sleep duration to obesity and T2DM and plausible mechanisms. In addition, we review the impact of changes in sleep duration on obesity and T2DM.
Collapse
Affiliation(s)
- Christina Antza
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Diabetes and Endocrinology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Georgios Kostopoulos
- Department of Endocrinology, 424 General Military Hospital, Thessaloniki, Greece
| | - Samiul Mostafa
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Diabetes and Endocrinology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Krishnarajah Nirantharakumar
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- Centre of Endocrinology Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Abd Tahrani
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Department of Diabetes and Endocrinology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Centre of Endocrinology Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Correspondence should be addressed to A Tahrani:
| |
Collapse
|
35
|
Yuan RK, Zitting KM, Duffy JF, Vujovic N, Wang W, Quan SF, Klerman EB, Scheer FAJL, Buxton OM, Williams JS, Czeisler CA. Chronic Sleep Restriction While Minimizing Circadian Disruption Does Not Adversely Affect Glucose Tolerance. Front Physiol 2021; 12:764737. [PMID: 34744800 PMCID: PMC8564292 DOI: 10.3389/fphys.2021.764737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/24/2021] [Indexed: 11/24/2022] Open
Abstract
Insufficient sleep, which has been shown to adversely affect metabolism, is generally associated with prolonged exposure to artificial light at night, a known circadian disruptor. There is growing evidence suggesting that circadian disruption adversely affects metabolism, yet few studies have attempted to evaluate the adverse metabolic effects of insufficient sleep while controlling for circadian disruption. We assessed postprandial glucose and insulin responses to a standard breakfast meal in healthy adults (n = 9) who underwent 3 weeks of chronic sleep restriction (CSR) in a 37-day inpatient study while minimizing circadian disruption by maintaining the same duration of light exposure each study day. We compared these results to findings from an earlier inpatient study which used a forced desynchrony (FD) protocol to assess the influence of 3 weeks of CSR combined with recurrent circadian disruption (RCD) on glycemic control in healthy adults (n = 21). CSR combined with RCD resulted in significantly elevated postprandial plasma glucose levels (p < 0.0001), while CSR with minimized circadian disruption had no adverse glycemic effects after 3 weeks of exposure (EXP). These results suggest that one mechanism by which sleep restriction impacts metabolism may be via concurrent circadian disruption.
Collapse
Affiliation(s)
- Robin K Yuan
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Kirsi-Marja Zitting
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Jeanne F Duffy
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Nina Vujovic
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Wei Wang
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Stuart F Quan
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Orfeu M Buxton
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States.,Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Charles A Czeisler
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, Boston, MA, United States.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
36
|
Abstract
Wearable technology has a history in sleep research dating back to the 1970s. Because modern wearable technology is relatively cheap and widely used by the general population, this represents an opportunity to leverage wearable devices to advance sleep medicine and research. However, there is a lack of published validation studies designed to quantify device performance against accepted gold standards, especially across different populations. Recommendations for conducting performance assessments and using wearable devices are now published with the goal of standardizing wearable device implementation and advancing the field.
Collapse
|
37
|
Moholdt T, Parr EB, Devlin BL, Debik J, Giskeødegård G, Hawley JA. The effect of morning vs evening exercise training on glycaemic control and serum metabolites in overweight/obese men: a randomised trial. Diabetologia 2021; 64:2061-2076. [PMID: 34009435 PMCID: PMC8382617 DOI: 10.1007/s00125-021-05477-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/09/2021] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS We determined whether the time of day of exercise training (morning vs evening) would modulate the effects of consumption of a high-fat diet (HFD) on glycaemic control, whole-body health markers and serum metabolomics. METHODS In this three-armed parallel-group randomised trial undertaken at a university in Melbourne, Australia, overweight/obese men consumed an HFD (65% of energy from fat) for 11 consecutive days. Participants were recruited via social media and community advertisements. Eligibility criteria for participation were male sex, age 30-45 years, BMI 27.0-35.0 kg/m2 and sedentary lifestyle. The main exclusion criteria were known CVD or type 2 diabetes, taking prescription medications, and shift-work. After 5 days, participants were allocated using a computer random generator to either exercise in the morning (06:30 hours), exercise in the evening (18:30 hours) or no exercise for the subsequent 5 days. Participants and researchers were not blinded to group assignment. Changes in serum metabolites, circulating lipids, cardiorespiratory fitness, BP, and glycaemic control (from continuous glucose monitoring) were compared between groups. RESULTS Twenty-five participants were randomised (morning exercise n = 9; evening exercise n = 8; no exercise n = 8) and 24 participants completed the study and were included in analyses (n = 8 per group). Five days of HFD induced marked perturbations in serum metabolites related to lipid and amino acid metabolism. Exercise training had a smaller impact than the HFD on changes in circulating metabolites, and only exercise undertaken in the evening was able to partly reverse some of the HFD-induced changes in metabolomic profiles. Twenty-four-hour glucose concentrations were lower after 5 days of HFD compared with the participants' habitual diet (5.3 ± 0.4 vs 5.6 ± 0.4 mmol/l, p = 0.001). There were no significant changes in 24 h glucose concentrations for either exercise group but lower nocturnal glucose levels were observed in participants who trained in the evening, compared with when they consumed the HFD alone (4.9 ± 0.4 vs 5.3 ± 0.3 mmol/l, p = 0.04). Compared with the no-exercise group, peak oxygen uptake improved after both morning (estimated effect 1.3 ml min-1 kg-1 [95% CI 0.5, 2.0], p = 0.003) and evening exercise (estimated effect 1.4 ml min-1 kg-1 [95% CI 0.6, 2.2], p = 0.001). Fasting blood glucose, insulin, cholesterol, triacylglycerol and LDL-cholesterol concentrations decreased only in participants allocated to evening exercise training. There were no unintended or adverse effects. CONCLUSIONS/INTERPRETATION A short-term HFD in overweight/obese men induced substantial alterations in lipid- and amino acid-related serum metabolites. Improvements in cardiorespiratory fitness were similar regardless of the time of day of exercise training. However, improvements in glycaemic control and partial reversal of HFD-induced changes in metabolic profiles were only observed when participants exercise trained in the evening. TRIAL REGISTRATION anzctr.org.au registration no. ACTRN12617000304336. FUNDING This study was funded by the Novo Nordisk Foundation (NNF14OC0011493).
Collapse
Affiliation(s)
- Trine Moholdt
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
- Women's Clinic, St Olavs Hospital, Trondheim, Norway.
| | - Evelyn B Parr
- Exercise & Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Fitzroy, VIC, Australia
| | - Brooke L Devlin
- Exercise & Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Fitzroy, VIC, Australia
- Department of Dietetics, Nutrition and Sport, La Trobe University, Melbourne, VIC, Australia
| | - Julia Debik
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Guro Giskeødegård
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - John A Hawley
- Exercise & Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Fitzroy, VIC, Australia.
| |
Collapse
|
38
|
Henderson GC. Plasma Free Fatty Acid Concentration as a Modifiable Risk Factor for Metabolic Disease. Nutrients 2021; 13:nu13082590. [PMID: 34444750 PMCID: PMC8402049 DOI: 10.3390/nu13082590] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/18/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Plasma free fatty acid (FFA) concentration is elevated in obesity, insulin resistance (IR), non-alcoholic fatty liver disease (NAFLD), type 2 diabetes (T2D), and related comorbidities such as cardiovascular disease (CVD). Furthermore, experimentally manipulating plasma FFA in the laboratory setting modulates metabolic markers of these disease processes. In this article, evidence is presented indicating that plasma FFA is a disease risk factor. Elevations of plasma FFA can promote ectopic lipid deposition, IR, as well as vascular and cardiac dysfunction. Typically, elevated plasma FFA results from accelerated adipose tissue lipolysis, caused by a high adipose tissue mass, adrenal hormones, or other physiological stressors. Reducing an individual’s postabsorptive and postprandial plasma FFA concentration is expected to improve health. Lifestyle change could provide a significant opportunity for plasma FFA reduction. Various factors can impact plasma FFA concentration, such as chronic restriction of dietary energy intake and weight loss, as well as exercise, sleep quality and quantity, and cigarette smoking. In this review, consideration is given to multiple factors which lead to plasma FFA elevation and subsequent disruption of metabolic health. From considering a variety of medical conditions and lifestyle factors, it becomes clear that plasma FFA concentration is a modifiable risk factor for metabolic disease.
Collapse
Affiliation(s)
- Gregory C Henderson
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
39
|
Prokopidis K, Dionyssiotis Y. Effects of sleep deprivation on sarcopenia and obesity: A narrative review of randomized controlled and crossover trials. J Frailty Sarcopenia Falls 2021; 6:50-56. [PMID: 34131601 PMCID: PMC8173530 DOI: 10.22540/jfsf-06-050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Shortened and fragmented sleeping patterns occupying modern industrialized societies may promote metabolic disturbances accompanied by increased risk of weight gain and skeletal muscle degradation. Short-term sleep restriction may alter energy homeostasis by modifying dopamine brain receptor signaling, leading to hyperpalatable food consumption and risk of increased adiposity. Concomitantly, the metabolic damage caused by lower testosterone and higher cortisol levels may stimulate systemic inflammation, insulin resistance, and suppress pathways involved in muscle protein synthesis. These changes may lead to dysregulated energy balance and skeletal muscle metabolism, increasing the risk of sarcopenic obesity, an additional public health burden. Future trials controlling for food intake and exploring further the influence of sleep deprivation on anabolic and catabolic signaling, and gut peptide interaction with energy balance are warranted.
Collapse
Affiliation(s)
- Konstantinos Prokopidis
- Imperial College London, Department of Metabolism, Digestion and Reproduction, London, United Kingdom
| | - Yannis Dionyssiotis
- 1 Physical Medicine and Rehabilitation Department, National Rehabilitation Center EKA, Athens, Greece
| |
Collapse
|
40
|
Porter JW, Pettit-Mee RJ, Emerson TS, McCrae CS, Lastra G, Vieira-Potter VJ, Parks EJ, Kanaley JA. Modest sleep restriction does not influence steps, physical activity intensity or glucose tolerance in obese adults. J Sleep Res 2021; 30:e13381. [PMID: 33949729 DOI: 10.1111/jsr.13381] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022]
Abstract
Sleep restriction (SR) (<6 h) and physical activity (PA) are risk factors for obesity, but little work has examined the inter-related influences of both risk factors. In a free-living environment, 13 overweight/obese adults were sleep restricted for five nights to 6 h time-in-bed each night, with and without regular exercise (45 min/65% VO2 max; counterbalanced design). Two days of recovery sleep followed SR. Subjects were measured during a mixed meal tolerance test (MMT), resting metabolic rate, cognitive testing and fat biopsy (n=8). SR increased peak glucose response (+7.3 mg/dl, p = .04), elevated fasting non-esterified fatty acid (NEFA) concentrations (+0.1 mmol/L, p = .001) and enhanced fat oxidation (p < .001) without modifying step counts or PA intensity. Inclusion of daily exercise increased step count (+4,700 steps/day, p < .001) and decreased the insulin response to a meal (p = .01) but did not prevent the increased peak glucose response or elevated NEFA levels. The weekend recovery period improved fasting glucose (p = .02), insulin (p = .02), NEFA concentrations (p = .001) and HOMA-IR (p < .01) despite reduced steps (p < .01) and increased sedentary time (p < .01). Abdominal adipose tissue (AT) samples, obtained after baseline, SR and exercise, did not differ in lipolytic capacity following SR. Fatty acid synthase protein content tended to increase following SR (p = .07), but not following exercise. In a free-living setting, SR adversely affected circulating NEFAs, fuel oxidation and peak glucose response but did not directly affect glucose tolerance or AT lipolysis. SR-associated metabolic impairments were not mitigated by exercise, yet recovery sleep completely rescued its adverse effects on glucose metabolism.
Collapse
Affiliation(s)
- Jay W Porter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Ryan J Pettit-Mee
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Travis S Emerson
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Christina S McCrae
- Department of Health Psychology, University of Missouri, Columbia, MO, USA
| | - Guido Lastra
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Missouri School of Medicine, Columbia, MO, USA
| | | | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
41
|
Al-Yousif N, Rawal S, Jurczak M, Mahmud H, Shah FA. Endogenous Glucose Production in Critical Illness. Nutr Clin Pract 2021; 36:344-359. [PMID: 33682953 DOI: 10.1002/ncp.10646] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regulation of endogenous glucose production (EGP) by hormonal, neuronal, and metabolic signaling pathways contributes to the maintenance of euglycemia under normal physiologic conditions. EGP is defined by the generation of glucose from substrates through glycogenolysis and gluconeogenesis, usually in fasted states, for local and systemic use. Abnormal increases in EGP are noted in patients with diabetes mellitus type 2, and elevated EGP may also impact the pathogenesis of nonalcoholic fatty liver disease and congestive heart failure. In this narrative review, we performed a literature search in PubMed to identify recently published English language articles characterizing EGP in critical illness. Evidence from preclinical and clinical studies demonstrates that critical illness can disrupt EGP through multiple mechanisms including increased systemic inflammation, counterregulatory hormone and catecholamine release, alterations in the hypothalamic-pituitary axis, insulin resistance, lactic acidosis, and iatrogenic insults such as vasopressors and glucocorticoids administered as part of clinical care. EGP contributes to hyperglycemia in critical illness when abnormally elevated and to hypoglycemia when abnormally depressed, each of which has been independently associated with increased mortality. Increased EGP may also promote protein catabolism that could worsen critical illness myopathy and impede recovery. Better understanding of the mechanisms and factors contributing to dysregulated EGP in critical illness may help in the development of therapeutic strategies that promote euglycemia, reduce intensive care unit-associated catabolism, and improve patient outcomes.
Collapse
Affiliation(s)
- Nameer Al-Yousif
- Department of Internal Medicine, UPMC Mercy Hospital, Pittsburgh, Pennsylvania, USA
| | - Sagar Rawal
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael Jurczak
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hussain Mahmud
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Faraaz Ali Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
- Acute Lung Injury Center of Excellence, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
42
|
Mosavat M, Mirsanjari M, Arabiat D, Smyth A, Whitehead L. The Role of Sleep Curtailment on Leptin Levels in Obesity and Diabetes Mellitus. Obes Facts 2021; 14:214-221. [PMID: 33756469 PMCID: PMC8138234 DOI: 10.1159/000514095] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence has identified sleep as a significant, but modifiable, risk factor for metabolic syndrome, diabetes, and obesity. Leptin, an adipocyte-derived peptide and a regulator of food intake and energy expenditure, has been shown to be associated with a short sleep duration in the pathophysiology of obesity and consequently type 2 diabetes. This review focuses on the current evidence indicating the effects of a short sleep duration on the regulation of leptin concentration in association with obesity and diabetes mellitus. In summary, the evidence suggests that sleep deprivation, by affecting leptin regulation, may lead to obesity and consequently development of type 2 diabetes through increased appetite and food intake. However, findings on the role of leptin in diabetes due to sleep deprivation are contradictory, and further studies with larger sample sizes are needed to confirm previous findings.
Collapse
Affiliation(s)
- Maryam Mosavat
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia,
| | - Mitra Mirsanjari
- Mazandaran University of Medical Sciences, Emam Khomeini Hospital, Mazandaran, Iran
| | - Diana Arabiat
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia
- Maternal and Child Nursing Department, The University of Jordan, Amman, Jordan
| | - Aisling Smyth
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia
| | - Lisa Whitehead
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia
| |
Collapse
|
43
|
Abstract
The endogenous timekeeping system evolved to anticipate the time of the day through the 24 hours cycle of the Earth's rotation. In mammals, the circadian clock governs rhythmic physiological and behavioral processes, including the daily oscillation in glucose metabolism, food intake, energy expenditure, and whole-body insulin sensitivity. The results from a series of studies have demonstrated that environmental or genetic alterations of the circadian cycle in humans and rodents are strongly associated with metabolic diseases such as obesity and type 2 diabetes. Emerging evidence suggests that astrocyte clocks have a crucial role in regulating molecular, physiological, and behavioral circadian rhythms such as glucose metabolism and insulin sensitivity. Given the concurrent high prevalence of type 2 diabetes and circadian disruption, understanding the mechanisms underlying glucose homeostasis regulation by the circadian clock and its dysregulation may improve glycemic control. In this review, we summarize the current knowledge on the tight interconnection between the timekeeping system, glucose homeostasis, and insulin sensitivity. We focus specifically on the involvement of astrocyte clocks, at the organism, cellular, and molecular levels, in the regulation of glucose metabolism.
Collapse
Affiliation(s)
- Olga Barca-Mayo
- Circadian and Glial Biology Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel López
- NeurObesity Lab, Physiology Department, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
44
|
Sweeney EL, Peart DJ, Ellis JG, Walshe IH. Impairments in glycaemic control do not increase linearly with repeated nights of sleep restriction in healthy adults: a randomised controlled trial. Appl Physiol Nutr Metab 2021; 46:1091-1096. [PMID: 33721507 DOI: 10.1139/apnm-2020-1025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Evidence suggests reduced glycaemic control following sleep restriction in healthy individuals. However, it remains unknown if impairments in glycaemic control increase with each additional night of sleep restriction in a linear manner. This randomised crossover study aimed to determine if the impairment in glycaemic control increases with each additional night of sleep restriction. Ten healthy individuals underwent 4 nights of control sleep (8 hours in bed) and 4 nights of sleep restriction (4 hours in bed) in a sleep laboratory. An oral glucose tolerance test was conducted each morning. Serum glucose and insulin were measured. Glucose and insulin area under the curve were higher overall in the sleep restriction trial compared with control (p < 0.001 and p = 0.033); however, no effect of day (p = 0.620 and p = 0.863) or interaction effect (p = 0.152 and p = 0.285) were observed. This supports previous literature showing a detrimental impact of sleep restriction on glucose regulation. The present findings, however, suggest the impairment in glycaemic control does not increase in a linear manner with an increasing number of nights of sleep restriction. This may have implications for the design of future studies examining sleep restriction and glycaemic control. Novelty: Four nights of sleep restriction impaired glycaemic control in healthy individuals, but did not do so in a linear manner. No effect of number of nights of restriction was found for glucose or insulin, which may have implications for future studies.
Collapse
Affiliation(s)
- Emma L Sweeney
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle Upon Tyne, NE1 8ST, United Kingdom.,School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, United Kingdom
| | - Daniel J Peart
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Jason G Ellis
- Northumbria Sleep Research Laboratory, Department of Psychology, Northumbria University, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Ian H Walshe
- Department of Sport, Exercise and Rehabilitation, Northumbria University, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| |
Collapse
|
45
|
Abstract
Polycystic ovarian syndrome (PCOS) is a common endocrinopathy with many clinical manifestations. The effects on women's lives start at puberty and can last throughout her lifetime. Women frequently experience anovulatory menstrual cycles, infertility, hirsutism, obesity and increased risk of diabetes mellitus, hypertension, lipid abnormalities, and metabolic syndrome. PCOS is a heterogenous disorder, and a diagnosis of exclusion. In general, women afflicted will have menstrual irregularities, ultrasound findings of abnormal ovarian size and morphology, and clinical or laboratory evidence of hyperandrogenism. This chapter reviews the current understanding of PCOS, associated metabolic abnormalities, and diagnosis in reproductive-aged women, as well as adolescents.
Collapse
Affiliation(s)
- Jessica A Lentscher
- Program of Reproductive Endocrinology and Infertility, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
46
|
Mikheev RK, Andreeva EN, Sheremetyeva EV, Absatarova YS, Ponomareva TA, Grigoryan OR. [Analysis of melatonin concentration and its correlation with ovarian disfunction among obese women of reproductive age]. ACTA ACUST UNITED AC 2021; 67:69-75. [PMID: 33586394 PMCID: PMC8926146 DOI: 10.14341/probl12710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/22/2021] [Indexed: 11/10/2022]
Abstract
Одним из новых направлений в изучении нарушений репродуктивной функции у женщин с ожирением являются влияние и рецепторная чувствительность мелатонина на гонадотропную функцию гипофиза и овариогенез с учетом хронологии «светового загрязнения». На современном этапе литературы по влиянию аспектов «светового загрязнения» на проблему ожирения и нарушений репродуктивной функции в отечественной и зарубежной литературе крайне мало. Данный обзор является попыткой объединить вышеуказанную проблему в рамках аспектов влияния «светового загрязнения» и уровня рецепторной чувствительности мелатонина у женщин репродуктивного возраста с ожирением. Поиск литературы проводили в отечественных (eLibrary, CyberLeninka.ru) и международных (PubMed, Cochrane Library) базах данных на русском и английском языках. Приоритетным являлся свободный доступ к полному тексту статей. Выбор источников был приоритетен периодом с 2015 по 2019 гг. Однако с учетом недостаточной изученности выбранной темы выбор источников датировался с 1992 г. Работа выполнена в рамках Государственного задания «Центральные и периферические патофизиологические механизмы развития болезней жировой ткани с учетом клинических и гормональных характеристик», 2020–2022 гг.
Collapse
Affiliation(s)
| | - E N Andreeva
- Endocrinology Research Centre; Moscow State University of Medicine and Dentistry
| | | | | | | | | |
Collapse
|
47
|
Mancilla R, Brouwers B, Schrauwen‐Hinderling VB, Hesselink MKC, Hoeks J, Schrauwen P. Exercise training elicits superior metabolic effects when performed in the afternoon compared to morning in metabolically compromised humans. Physiol Rep 2021; 8:e14669. [PMID: 33356015 PMCID: PMC7757369 DOI: 10.14814/phy2.14669] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/13/2020] [Accepted: 11/14/2020] [Indexed: 01/08/2023] Open
Abstract
The circadian clock and metabolism are tightly intertwined. Hence, the specific timing of interventions that target metabolic changes may affect their efficacy. Here we retrospectively compared the metabolic health effects of morning versus afternoon exercise training in metabolically compromised subjects enrolled in a 12-week exercise training program. Thirty-two adult males (58 ± 7 yrs) at risk for or diagnosed with type 2 diabetes performed 12 weeks of supervised exercise training either in the morning (8.00-10.00 a.m., N = 12) or in the afternoon (3.00-6.00 p.m., N = 20). Compared to participants who trained in the morning, participants who trained in the afternoon experienced superior beneficial effects of exercise training on peripheral insulin sensitivity (+5.2 ± 6.4 vs. -0.5 ± 5.4 μmol/min/kgFFM, p = .03), insulin-mediated suppression of adipose tissue lipolysis (-4.5 ± 13.7% vs. +5.9 ± 11%, p = .04), fasting plasma glucose levels (-0.3 ± 1.0 vs. +0.5 ± 0.8 mmol/l, p = .02), exercise performance (+0.40 ± 0.2 vs. +0.2 ± 0.1 W/kg, p = .05) and fat mass (-1.2 ± 1.3 vs. -0.2 ± 1.0 kg, p = .03). In addition, exercise training in the afternoon also tended to elicit superior effects on basal hepatic glucose output (p = .057). Our findings suggest that metabolically compromised subjects may reap more pronounced metabolic benefits from exercise training when this training is performed in the afternoon versus morning. CLINICALTRIALS.GOV ID: NCT01317576.
Collapse
Affiliation(s)
- Rodrigo Mancilla
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Nutrition and Movement SciencesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Bram Brouwers
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Nutrition and Movement SciencesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Vera B. Schrauwen‐Hinderling
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of RadiologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Matthijs K. C. Hesselink
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Nutrition and Movement SciencesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Joris Hoeks
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Nutrition and Movement SciencesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Patrick Schrauwen
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of Nutrition and Movement SciencesMaastricht University Medical CenterMaastrichtThe Netherlands
| |
Collapse
|
48
|
Saner NJ, Lee MJC, Kuang J, Pitchford NW, Roach GD, Garnham A, Genders AJ, Stokes T, Schroder EA, Huo Z, Esser KA, Phillips SM, Bishop DJ, Bartlett JD. Exercise mitigates sleep-loss-induced changes in glucose tolerance, mitochondrial function, sarcoplasmic protein synthesis, and diurnal rhythms. Mol Metab 2021; 43:101110. [PMID: 33137489 PMCID: PMC7704425 DOI: 10.1016/j.molmet.2020.101110] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Sleep loss has emerged as a risk factor for the development of impaired glucose tolerance. The mechanisms underpinning this observation are unknown; however, both mitochondrial dysfunction and circadian misalignment have been proposed. Because exercise improves glucose tolerance and mitochondrial function, and alters circadian rhythms, we investigated whether exercise may counteract the effects induced by inadequate sleep. METHODS To minimize between-group differences of baseline characteristics, 24 healthy young males were allocated into one of the three experimental groups: a Normal Sleep (NS) group (8 h time in bed (TIB) per night, for five nights), a Sleep Restriction (SR) group (4 h TIB per night, for five nights), and a Sleep Restriction and Exercise group (SR+EX) (4 h TIB per night, for five nights and three high-intensity interval exercise (HIIE) sessions). Glucose tolerance, mitochondrial respiratory function, sarcoplasmic protein synthesis (SarcPS), and diurnal measures of peripheral skin temperature were assessed pre- and post-intervention. RESULTS We report that the SR group had reduced glucose tolerance post-intervention (mean change ± SD, P value, SR glucose AUC: 149 ± 115 A.U., P = 0.002), which was also associated with reductions in mitochondrial respiratory function (SR: -15.9 ± 12.4 pmol O2.s-1.mg-1, P = 0.001), a lower rate of SarcPS (FSR%/day SR: 1.11 ± 0.25%, P < 0.001), and reduced amplitude of diurnal rhythms. These effects were not observed when incorporating three sessions of HIIE during this period (SR+EX: glucose AUC 67 ± 57, P = 0.239, mitochondrial respiratory function: 0.6 ± 11.8 pmol O2.s-1.mg-1, P = 0.997, and SarcPS (FSR%/day): 1.77 ± 0.22%, P = 0.971). CONCLUSIONS A five-night period of sleep restriction leads to reductions in mitochondrial respiratory function, SarcPS, and amplitude of skin temperature diurnal rhythms, with a concurrent reduction in glucose tolerance. We provide novel data demonstrating that these same detrimental effects are not observed when HIIE is performed during the period of sleep restriction. These data therefore provide evidence in support of the use of HIIE as an intervention to mitigate the detrimental physiological effects of sleep loss.
Collapse
Affiliation(s)
- Nicholas J Saner
- Institute for Health and Sport, Victoria University, Melbourne, Australia; Sports Cardiology, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Matthew J-C Lee
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Jujiao Kuang
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Nathan W Pitchford
- Institute for Health and Sport, Victoria University, Melbourne, Australia; Sport Performance Optimization Research Team, School of Human Life Sciences, University of Tasmania, Launceston, Australia
| | - Gregory D Roach
- Appleton Institute for Behavioral Science, Central Queensland University, Adelaide, Australia
| | - Andrew Garnham
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Amanda J Genders
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Tanner Stokes
- Department of Kinesiology, McMaster University, Hamilton, Canada
| | - Elizabeth A Schroder
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, United States
| | - Zhiguang Huo
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, United States
| | | | - David J Bishop
- Institute for Health and Sport, Victoria University, Melbourne, Australia.
| | | |
Collapse
|
49
|
Brzecka A, Madetko N, Nikolenko VN, Ashraf GM, Ejma M, Leszek J, Daroszewski C, Sarul K, Mikhaleva LM, Somasundaram SG, Kirkland CE, Bachurin SO, Aliev G. Sleep Disturbances and Cognitive Impairment in the Course of Type 2 Diabetes-A Possible Link. Curr Neuropharmacol 2020; 19:78-91. [PMID: 32148197 PMCID: PMC7903492 DOI: 10.2174/1570159x18666200309101750] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
There is an increasing number of patients worldwide with sleep disturbances and diabetes. Various sleep disorders, including long or short sleep duration and poor sleep quality of numerous causes, may increase the risk of diabetes. Some symptoms of diabetes, such as painful peripheral neuropathy and nocturia, or associated other sleep disorders, such as sleep breathing disorders or sleep movement disorders, may influence sleep quality and quantity. Both sleep disorders and diabetes may lead to cognitive impairment. The risk of development of cognitive impairment in diabetic patients may be related to vascular and non-vascular and other factors, such as hypoglycemia, hyperglycemia, central insulin resistance, amyloid and tau deposits and other causes. Numerous sleep disorders, e.g., sleep apnea, restless legs syndrome, insomnia, and poor sleep quality are most likely are also associated with cognitive impairment. Adequate functioning of the system of clearance of the brain from toxic substances, such as amyloid β, i.e. glymphatic system, is related to undisturbed sleep and prevents cognitive impairment. In the case of coexistence, sleep disturbances and diabetes either independently lead to and/or mutually aggravate cognitive impairment.
Collapse
Affiliation(s)
- Anna Brzecka
- Department of Pulmonology and Lung Cancer, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Madetko
- Department of Neurology, Wroclaw Medical University, Wroclaw, Poland
| | - Vladimir N Nikolenko
- I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russian Federation
| | - Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maria Ejma
- Department of Neurology, Wroclaw Medical University, Wroclaw, Poland
| | - Jerzy Leszek
- Department of Psychiatry, Wroclaw Medical University, Wroclaw, Poland
| | - Cyryl Daroszewski
- Department of Pulmonology and Lung Cancer, Wroclaw Medical University, Wroclaw, Poland
| | - Karolina Sarul
- Department of Pulmonology and Lung Cancer, Wroclaw Medical University, Wroclaw, Poland
| | - Liudmila M Mikhaleva
- Research Institute of Human Morphology,3 Tsyurupy Street, Moscow, 117418, Russian Federation
| | - Siva G Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV, 26426, United States
| | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, Salem, WV, 26426, United States
| | - Sergey O Bachurin
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
| | - Gjumrakch Aliev
- I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russian Federation
| |
Collapse
|
50
|
Gao X, Sun H, Zhang Y, Liu L, Wang J, Wang T. Investigating Causal Relations Between Sleep-Related Traits and Risk of Type 2 Diabetes Mellitus: A Mendelian Randomization Study. Front Genet 2020; 11:607865. [PMID: 33384720 PMCID: PMC7770175 DOI: 10.3389/fgene.2020.607865] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Objective Extensive literature put forward the link between sleep and type 2 diabetes mellitus (T2DM), however, little is known about the underlying causality of the associations. Here we aim to assess the causal relationships between five major sleep-related traits and T2DM. Design, Setting, and Participants Two-sample Mendelian randomization (MR) was utilized to investigate the potential causal relations. Independent genetic variants associated with five sleep-related phenotypes—insomnia, sleep duration, short sleep duration, long sleep duration, and morningness—were chosen as instrumental variables to estimate the causal associations with T2DM. Summary statistics were acquired from the genome-wide association studies of UK Biobank and 23andMe (for sleep-related measures), the DIAbetes Genetics Replication And Meta-analysis and the FinnGen (for T2DM). Main Methods Individual Cochran’s Q statistic was applied to remove the pleiotropic instruments, global Q statistics and MR-Egger regression were adopted to test for the global heterogeneity and horizontal pleiotropy of the screened instruments, respectively. Two T2DM cohorts were selected to analyze their associations with sleep traits. A modified inverse variance weighted (IVW) estimate was performed to combine the ratio estimators from each instrument and acquire the causal estimate, alternative methods including IVW with first-order weights, simple and weighted median estimations, and MR-Egger regression were conducted as sensitivity analyses, to ensure the robustness and solidity of the findings. Results Two-sample MR supported findings for an adverse effect of genetically predicted insomnia on T2DM risk (odds ratio [OR] = 1.14, 95% confidence interval [CI]: 1.09–1.19, p = 1.29E–08) at the Bonferroni-adjusted level of significance (p < 0.005). We further investigated the causal role of T2DM on insomnia but obtained a non-significant estimation. There was also little evidence for the causal effect of other sleep-related measures on T2DM. Results were largely consistent when leveraging two different T2DM cohorts, and were robust among various sensitivity analyses. Conclusion Findings provide significant evidence for an adverse effect of insomnia on T2DM risk. The study extends fundamental knowledge to further understanding of the pathophysiological mechanisms of T2DM, and points out the non-negligible role of insomnia on epidemiologic intervention and clinical therapeutics of T2DM.
Collapse
Affiliation(s)
- Xue Gao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Heli Sun
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yu Zhang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Long Liu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Juping Wang
- Department of Mathematics, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| |
Collapse
|