1
|
Jakubowicz D, Matz Y, Landau Z, Rosenblum RC, Twito O, Wainstein J, Tsameret S. Interaction Between Early Meals (Big-Breakfast Diet), Clock Gene mRNA Expression, and Gut Microbiome to Regulate Weight Loss and Glucose Metabolism in Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:12355. [PMID: 39596418 PMCID: PMC11594859 DOI: 10.3390/ijms252212355] [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: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The circadian clock gene system plays a pivotal role in coordinating the daily rhythms of most metabolic processes. It is synchronized with the light-dark cycle and the eating-fasting schedule. Notably, the interaction between meal timing and circadian clock genes (CGs) allows for optimizing metabolic processes at specific times of the day. Breakfast has a powerful resetting effect on the CG network. A misaligned meal pattern, such as skipping breakfast, can lead to a discordance between meal timing and the endogenous CGs, and is associated with obesity and T2D. Conversely, concentrating most calories and carbohydrates (CH) in the early hours of the day upregulates metabolic CG expression, thus promoting improved weight loss and glycemic control. Recently, it was revealed that microorganisms in the gastrointestinal tract, known as the gut microbiome (GM), and its derived metabolites display daily oscillation, and play a critical role in energy and glucose metabolism. The timing of meal intake coordinates the oscillation of GM and GM-derived metabolites, which in turn influences CG expression, playing a crucial role in the metabolic response to food intake. An imbalance in the gut microbiota (dysbiosis) can also reciprocally disrupt CG rhythms. Evidence suggests that misaligned meal timing may cause such disruptions and can lead to obesity and hyperglycemia. This manuscript focuses on the reciprocal interaction between meal timing, GM oscillation, and circadian CG rhythms. It will also review studies demonstrating how aligning meal timing with the circadian clock can reset and synchronize CG rhythms and GM oscillations. This synchronization can facilitate weight loss and improve glycemic control in obesity and those with T2D.
Collapse
Affiliation(s)
- Daniela Jakubowicz
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Yael Matz
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Zohar Landau
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Rachel Chava Rosenblum
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Orit Twito
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Julio Wainstein
- Endocrinology and Diabetes Unit, Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon 58100, Israel
| | - Shani Tsameret
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
2
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
3
|
Daniels LJ, Kay D, Marjot T, Hodson L, Ray DW. Circadian regulation of liver metabolism: experimental approaches in human, rodent, and cellular models. Am J Physiol Cell Physiol 2023; 325:C1158-C1177. [PMID: 37642240 PMCID: PMC10861179 DOI: 10.1152/ajpcell.00551.2022] [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: 12/19/2022] [Revised: 06/15/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023]
Abstract
Circadian rhythms are endogenous oscillations with approximately a 24-h period that allow organisms to anticipate the change between day and night. Disruptions that desynchronize or misalign circadian rhythms are associated with an increased risk of cardiometabolic disease. This review focuses on the liver circadian clock as relevant to the risk of developing metabolic diseases including nonalcoholic fatty liver disease (NAFLD), insulin resistance, and type 2 diabetes (T2D). Many liver functions exhibit rhythmicity. Approximately 40% of the hepatic transcriptome exhibits 24-h rhythms, along with rhythms in protein levels, posttranslational modification, and various metabolites. The liver circadian clock is critical for maintaining glucose and lipid homeostasis. Most of the attention in the metabolic field has been directed toward diet, exercise, and rather little to modifiable risks due to circadian misalignment or disruption. Therefore, the aim of this review is to systematically analyze the various approaches that study liver circadian pathways, targeting metabolic liver diseases, such as diabetes, nonalcoholic fatty liver disease, using human, rodent, and cell biology models.NEW & NOTEWORTHY Over the past decade, there has been an increased interest in understanding the intricate relationship between circadian rhythm and liver metabolism. In this review, we have systematically searched the literature to analyze the various experimental approaches utilizing human, rodent, and in vitro cellular approaches to dissect the link between liver circadian rhythms and metabolic disease.
Collapse
Affiliation(s)
- Lorna J Daniels
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Danielle Kay
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
- Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Scott EM, Murphy HR, Myers J, Saravanan P, Poston L, Law GR. MAGIC (maternal glucose in pregnancy) understanding the glycemic profile of pregnancy, intensive CGM glucose profiling and its relationship to fetal growth: an observational study protocol. BMC Pregnancy Childbirth 2023; 23:563. [PMID: 37537535 PMCID: PMC10398923 DOI: 10.1186/s12884-023-05824-x] [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: 03/27/2023] [Accepted: 06/30/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Continuous glucose monitoring (CGM) provides the most objective method of assessing glucose in daily life. Although there have been small, short-term physiologic studies of glucose metabolism in 'healthy' pregnant women a comprehensive, longitudinal description of changes in glucose over the course of pregnancy and how glucose dysregulation earlier in pregnancy relates to traditional third trimester screening for gestational diabetes, fetal growth and pregnancy outcomes is lacking. This study aims to characterise longitudinal changes in glycemia across gestation using CGM, in order to understand the evolution of dysglycemia and its relationship to fetal growth. METHOD/DESIGN A multi-centre, prospective, observational, cohort study of 500 healthy pregnant women, recruited in the first trimester of pregnancy. Masked CGM will be performed for a 14-day period on five occasions across pregnancy at ~ 10-12, 18-20, 26-28, 34-36 weeks gestation and postnatally. Routinely collected anthropometric and sociodemographic information will be recorded at each visit including: weight, height, blood pressure, current medication. Age, parity, ethnicity, smoking will be recorded. Blood samples will be taken at each visit for HbA1c and a sample stored. Details on fetal growth from ultrasound scans and the OGTT results will be recorded. Maternal and neonatal outcomes will be collected. CGM glucose profiling is the exposure of interest, and will be performed using standard summary statistics, functional data analysis and glucotyping. The primary maternal outcome is clinical diagnosis of GDM. The primary neonatal outcome is large for gestational age (LGA) (> 90th centile defined by customised birthweight centile). The relationship of glucose to key secondary maternal and neonatal outcomes will be explored. DISCUSSION This study will ascertain the relationship of maternal dysglycemia to fetal growth and outcomes. It will explore whether CGM glucose profiling can detect GDM before the OGTT; or indeed whether CGM glucose profiling may be more useful than the OGTT at detecting LGA and other perinatal outcomes. TRIAL REGISTRATION ISRCTN 15,706,303 https://www.isrctn.com/ISRCTN15706303 Registration date: 13th March 2023.
Collapse
Affiliation(s)
- Eleanor M Scott
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, LIGHT Laboratories, University of Leeds, Clarendon Way, Leeds, LS2 9JT, UK.
| | - Helen R Murphy
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Ponnusamy Saravanan
- Populations, Evidence and Technologies, Division of Health Sciences, Warwick Medical School, University of Warwick, Warwick, UK
- Diabetes, Endocrinology and Metabolism, George Eliot Hospital, Nuneaton, UK
| | - Lucilla Poston
- Tommy's Maternal and Fetal Research Unit, Kings College London, London, UK
| | | |
Collapse
|
5
|
Petrecca S, Quail DF. Mouse Models of Obesity to Study the Tumor-Immune Microenvironment. Methods Mol Biol 2023; 2614:121-138. [PMID: 36587123 DOI: 10.1007/978-1-0716-2914-7_9] [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: 01/02/2023]
Abstract
Obesity is associated with chronic, low-grade systemic inflammation and leads to changes in the immune microenvironment of various tissues. As a result, obesity is associated with increased risk of cancer and a worse prognosis in patients. Given the prevalence of obesity worldwide, understanding the fundamental biology governing the relationship between obesity and cancer is critical. In this chapter, we describe preclinical models of obesity that can be combined with standard tumor models and techniques to study the tumor-immune microenvironment. We also discuss important considerations when planning experiments involving these models.
Collapse
Affiliation(s)
- Sarah Petrecca
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.,Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada. .,Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada. .,Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Downton P, Sanna F, Maidstone R, Poolman TM, Hayter EA, Dickson SH, Ciccone NA, Early JO, Adamson A, Spiller DG, Simpkins DA, Baxter M, Fischer R, Rattray M, Loudon ASI, Gibbs JE, Bechtold DA, Ray DW. Chronic inflammatory arthritis drives systemic changes in circadian energy metabolism. Proc Natl Acad Sci U S A 2022; 119:e2112781119. [PMID: 35482925 PMCID: PMC9170023 DOI: 10.1073/pnas.2112781119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 03/01/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic inflammation underpins many human diseases. Morbidity and mortality associated with chronic inflammation are often mediated through metabolic dysfunction. Inflammatory and metabolic processes vary through circadian time, suggesting an important temporal crosstalk between these systems. Using an established mouse model of rheumatoid arthritis, we show that chronic inflammatory arthritis results in rhythmic joint inflammation and drives major changes in muscle and liver energy metabolism and rhythmic gene expression. Transcriptional and phosphoproteomic analyses revealed alterations in lipid metabolism and mitochondrial function associated with increased EGFR-JAK-STAT3 signaling. Metabolomic analyses confirmed rhythmic metabolic rewiring with impaired β-oxidation and lipid handling and revealed a pronounced shunt toward sphingolipid and ceramide accumulation. The arthritis-related production of ceramides was most pronounced during the day, which is the time of peak inflammation and increased reliance on fatty acid oxidation. Thus, our data demonstrate that localized joint inflammation drives a time-of-day–dependent build-up of bioactive lipid species driven by rhythmic inflammation and altered EGFR-STAT signaling.
Collapse
Affiliation(s)
- Polly Downton
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Fabio Sanna
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Robert Maidstone
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Toryn M. Poolman
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Edward A. Hayter
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Suzanna H. Dickson
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Nick A. Ciccone
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - James O. Early
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Antony Adamson
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David G. Spiller
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Devin A. Simpkins
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Matthew Baxter
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, OX3 7FZ, United Kingdom
| | - Magnus Rattray
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Andrew S. I. Loudon
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Julie E. Gibbs
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David A. Bechtold
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - David W. Ray
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
7
|
Martchenko SE, Prescott D, Martchenko A, Sweeney ME, Philpott DJ, Brubaker PL. Diurnal changes in the murine small intestine are disrupted by obesogenic Western Diet feeding and microbial dysbiosis. Sci Rep 2021; 11:20571. [PMID: 34663882 PMCID: PMC8523685 DOI: 10.1038/s41598-021-98986-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Intestinal functions demonstrate circadian rhythms thought to be entrained, in part, by an organisms’ intrinsic feeding and fasting periods as well as by the intestinal microbiome. Circadian disruption as a result of ill-timed nutrient exposure and obesogenic feeding poses an increased risk to disease. As such, the aim of this study was to assess the relationships between dietary timing, composition, and the microbiome with regard to rhythmic small intestinal structure and mucosal immunity. Rodent chow (RC)-mice exhibited time-dependent increases in small intestinal weight, villus height, and crypt depth as well as an increased proportion of CD8αα+ cells and concomitant decrease in CD8αβ+ cells at the onset of the feeding period (p < 0.05–0.001). Western diet (WD)-animals displayed disrupted time-dependent patterns in intestinal structure and lymphocyte populations (p < 0.05–0.01). Antibiotic-induced microbial depletion abrogated the time- and diet-dependent patterns in both RC- and WD-mice (p < 0.05–0.001). However, although germ-free-mice displayed altered rhythms, fecal microbial transfer from RC-mice was generally unsuccessful in restoring structural and immune changes in these animals. This study shows that adaptive changes in the small intestine at the onset of the feeding and fasting periods are disrupted by WD-feeding, and that these changes are dependent, in part, on the intestinal microbiome.
Collapse
Affiliation(s)
- Sarah E Martchenko
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - David Prescott
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Alexandre Martchenko
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Maegan E Sweeney
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Patricia L Brubaker
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Oladele CA, Akintayo CO, Badejogbin OC, Oniyide AA, Omoaghe AO, Agunbiade TB, Olaniyi KS. Melatonin ameliorates endocrine dysfunction and defective sperm integrity associated with high-fat diet-induced obesity in male Wistar rats. Andrologia 2021; 54:e14242. [PMID: 34490912 DOI: 10.1111/and.14242] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/12/2021] [Accepted: 08/31/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity (OBS) has been established as a link to male hypogonadism with consequent infertility. Previous studies have shown that melatonin (MEL) modulates hypothalamic-pituitary-gonadal function. The present study therefore investigated the hypothesis that MEL supplementation would attenuate spermatogenic and steroidogenic dysfunctions associated with obesity induced by high-fat diet (HFD). Twenty-four adult male Wistar rats (n = 6/group) were used: control group received vehicle (normal saline), obese group received 40% high-fat diet and distilled water, MEL-treated group received MEL (4 mg/kg), and OBS + MEL group received MEL and 40% HFD and the treatment lasted for 12 weeks. HFD caused increased body weight, glucose intolerance, plasma triglyceride and low-density lipoprotein cholesterol/ very low-density lipoprotein cholesterol and malondialdehyde, as well as decreased antioxidant capacity, high-density lipoprotein cholesterol, gonadotrophin-releasing hormone, follicle-stimulating hormone and testosterone and altered sperm parameters. However, all these alterations were attenuated when supplemented with MEL. Taken together, these results indicate that HFD exposure causes endocrine dysfunction and disrupted sperm parameters in obese animals, which are accompanied by lipid peroxidation/defective antioxidant capacity. In addition, the present results suggest that melatonin supplementation restores endocrine function and sperm integrity in obese rat model by suppression of oxidative stress-dependent mechanism.
Collapse
Affiliation(s)
- Comfort Abisola Oladele
- Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | | | | | - Adesola Adedotun Oniyide
- Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Adams Olalekan Omoaghe
- Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Toluwani Bosede Agunbiade
- Department of Medical Microbiology and Parasitology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Kehinde Samuel Olaniyi
- Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| |
Collapse
|
9
|
Role of High Energy Breakfast "Big Breakfast Diet" in Clock Gene Regulation of Postprandial Hyperglycemia and Weight Loss in Type 2 Diabetes. Nutrients 2021; 13:nu13051558. [PMID: 34063109 PMCID: PMC8148179 DOI: 10.3390/nu13051558] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 01/15/2023] Open
Abstract
Postprandial hyperglycemia (PPHG) is strongly linked with the future development of cardiovascular complications in type 2 diabetes (T2D). Hence, reducing postprandial glycemic excursions is essential in T2D treatment to slow progressive deficiency of β-cell function and prevent cardiovascular complications. Most of the metabolic processes involved in PPHG, i.e., β-cell secretory function, GLP-1 secretion, insulin sensitivity, muscular glucose uptake, and hepatic glucose production, are controlled by the circadian clock and display daily oscillation. Consequently, postprandial glycemia displays diurnal variation with a higher glycemic response after meals with the same carbohydrate content, consumed at dusk compared to the morning. T2D and meal timing schedule not synchronized with the circadian clock (i.e., skipping breakfast) are associated with disrupted clock gene expression and is linked to PPHG. In contrast, greater intake in the morning (i.e., high energy breakfast) than in the evening has a resetting effect on clock gene oscillations and beneficial effects on weight loss, appetite, and reduction of PPHG, independently of total energy intake. Therefore, resetting clock gene expression through a diet intervention consisting of meal timing aligned to the circadian clock, i.e., shifting most calories and carbohydrates to the early hours of the day, is a promising therapeutic approach to improve PPHG in T2D. This review will focus on recent studies, showing how a high-energy breakfast diet (Bdiet) has resetting and synchronizing actions on circadian clock genes expression, improving glucose metabolism, postprandial glycemic excursions along with weight loss in T2D.
Collapse
|
10
|
Lo EH, Albers GW, Dichgans M, Donnan G, Esposito E, Foster R, Howells DW, Huang YG, Ji X, Klerman EB, Lee S, Li W, Liebeskind DS, Lizasoain I, Mandeville ET, Moro MA, Ning M, Ray D, Sakadžić S, Saver JL, Scheer FAJL, Selim M, Tiedt S, Zhang F, Buchan AM. Circadian Biology and Stroke. Stroke 2021; 52:2180-2190. [PMID: 33940951 DOI: 10.1161/strokeaha.120.031742] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Circadian biology modulates almost all aspects of mammalian physiology, disease, and response to therapies. Emerging data suggest that circadian biology may significantly affect the mechanisms of susceptibility, injury, recovery, and the response to therapy in stroke. In this review/perspective, we survey the accumulating literature and attempt to connect molecular, cellular, and physiological pathways in circadian biology to clinical consequences in stroke. Accounting for the complex and multifactorial effects of circadian rhythm may improve translational opportunities for stroke diagnostics and therapeutics.
Collapse
Affiliation(s)
- Eng H Lo
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Gregory W Albers
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Stanford Stroke Center, Stanford University, Palo Alto (G.W.A., S.L.)
| | - Martin Dichgans
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,German Center for Neurodegenerative Diseases (DZNE, Munich) and Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.).,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M.D., S.T.)
| | - Geoffrey Donnan
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Medicine and Neurology, Royal Melbourne Hospital, University of Melbourne, Australia (G.D.)
| | - Elga Esposito
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Russell Foster
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences (R.F.), University of Oxford, United Kingdom
| | - David W Howells
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Tasmanian School of Medicine, University of Tasmania, Australia (D.W.H.)
| | - Yi-Ge Huang
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Stroke Medicine (Y.H., A.M.B.), University of Oxford, United Kingdom
| | - Xunming Ji
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Beijing Institute for Brain Disorders, China (X.J.)
| | - Elizabeth B Klerman
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Neurology (E.B.K., M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sarah Lee
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Stanford Stroke Center, Stanford University, Palo Alto (G.W.A., S.L.)
| | - Wenlu Li
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - David S Liebeskind
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Geffen School of Medicine, University of California Los Angeles (J.L.S., D.S.L.)
| | - Ignacio Lizasoain
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Pharmacology and Toxicology, Complutense Medical School, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain (I.L.)
| | - Emiri T Mandeville
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Maria A Moro
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain (M.A.M.)
| | - MingMing Ning
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Neurology (E.B.K., M.N.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - David Ray
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, and Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, United Kingdom (D.R.)
| | - Sava Sakadžić
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jeffrey L Saver
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Geffen School of Medicine, University of California Los Angeles (J.L.S., D.S.L.)
| | - Frank A J L Scheer
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Medicine and Neurology, Brigham & Women's Hospital (F.A.J.L.S.), Harvard Medical School, Boston
| | - Magdy Selim
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Beth Israel Deaconess Medical Center (M.S.), Harvard Medical School, Boston
| | - Steffen Tiedt
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M.D., S.T.)
| | - Fang Zhang
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Departments of Radiology (E.H.L., E.E., W.L., E.T.M., S.S., F.Z.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Alastair M Buchan
- CIRCA consortium (E.H.L., G.W.A., M.D., G.D., E.E., R.F., D.W.H., Y-G.H., X.J., E.B.K., S.L., W.L., D.S.L., I.L., E.T.M., M.A.M., M.N., D.R., S.S., J.L.S., F.A.J.L.S., M.S., S.T., F.Z., A.M.B.), Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Stroke Medicine (Y.H., A.M.B.), University of Oxford, United Kingdom
| |
Collapse
|
11
|
Maury E, Navez B, Brichard SM. Circadian clock dysfunction in human omental fat links obesity to metabolic inflammation. Nat Commun 2021; 12:2388. [PMID: 33888702 PMCID: PMC8062496 DOI: 10.1038/s41467-021-22571-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 03/12/2021] [Indexed: 01/01/2023] Open
Abstract
To unravel the pathogenesis of obesity and its complications, we investigate the interplay between circadian clocks and NF-κB pathway in human adipose tissue. The circadian clock function is impaired in omental fat from obese patients. ChIP-seq analyses reveal that the core clock activator, BMAL1 binds to several thousand target genes. NF-κB competes with BMAL1 for transcriptional control of some targets and overall, BMAL1 chromatin binding occurs in close proximity to NF-κB consensus motifs. Obesity relocalizes BMAL1 occupancy genome-wide in human omental fat, thereby altering the transcription of numerous target genes involved in metabolic inflammation and adipose tissue remodeling. Eventually, clock dysfunction appears at early stages of obesity in mice and is corrected, together with impaired metabolism, by NF-κB inhibition. Collectively, our results reveal a relationship between NF-κB and the molecular clock in adipose tissue, which may contribute to obesity-related complications.
Collapse
Affiliation(s)
- Eleonore Maury
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium.
| | - Benoit Navez
- Digestive Surgery Unit, Saint-Luc University Hospital, UCLouvain, Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| |
Collapse
|
12
|
"Shedding Light on Light": A Review on the Effects on Mental Health of Exposure to Optical Radiation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18041670. [PMID: 33572423 PMCID: PMC7916252 DOI: 10.3390/ijerph18041670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/11/2021] [Accepted: 02/03/2021] [Indexed: 01/10/2023]
Abstract
In relation to human health and functioning, light, or more specifically optical radiation, plays many roles, beyond allowing vision. These may be summarized as: regulation of circadian rhythms; consequences of direct exposure to the skin; and more indirect effects on well-being and functioning, also related to lifestyle and contact with natural and urban environments. Impact on mental health is relevant for any of these specifications and supports a clinical use of this knowledge for the treatment of psychiatric conditions, such as depression or anxiety, somatic symptom disorder, and others, with reference to light therapy in particular. The scope of this narrative review is to provide a summary of recent findings and evidence on the regulating functions of light on human beings’ biology, with a specific focus on mental health, its prevention and care.
Collapse
|
13
|
Scott EM, Feig DS, Murphy HR, Law GR. Continuous Glucose Monitoring in Pregnancy: Importance of Analyzing Temporal Profiles to Understand Clinical Outcomes. Diabetes Care 2020; 43:1178-1184. [PMID: 32209645 PMCID: PMC7245356 DOI: 10.2337/dc19-2527] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/28/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine if temporal glucose profiles differed between 1) women who were randomized to real-time continuous glucose monitoring (RT-CGM) or self-monitored blood glucose (SMBG), 2) women who used insulin pumps or multiple daily insulin injections (MDIs), and 3) women whose infants were born large for gestational age (LGA) or not, by assessing CGM data obtained from the Continuous Glucose Monitoring in Women With Type 1 Diabetes in Pregnancy Trial (CONCEPTT). RESEARCH DESIGN AND METHODS Standard summary metrics and functional data analysis (FDA) were applied to CGM data from the CONCEPTT trial (RT-CGM, n = 100; SMBG, n = 100) taken at baseline and at 24- and 34-weeks' gestation. Multivariable regression analysis determined if temporal differences in 24-h glucose profiles occurred between comparators in each of the three groups. RESULTS FDA revealed that women using RT-CGM had significantly lower glucose (0.4-0.8 mmol/L [7-14 mg/dL]) for 7 h/day (0800 h to 1200 h and 1600 h to 1900 h) compared with those with SMBG. Women using pumps had significantly higher glucose (0.4-0.9 mmol/L [7-16 mg/dL]) for 12 h/day (0300 h to 0600 h, 1300 h to 1800 h, and 2030 h to 0030 h) at 24 weeks with no difference at 34 weeks compared with MDI. Women who had an LGA infant ran a significantly higher glucose by 0.4-0.7 mmol/L (7-13 mg/dL) for 4.5 h/day at baseline, by 0.4-0.9 mmol/L (7-16 mg/dL) for 16 h/day at 24 weeks, and by 0.4-0.7 mmol/L (7-13 mg/dL) for 14 h/day at 34 weeks. CONCLUSIONS FDA of temporal glucose profiles gives important information about differences in glucose control and its timing, which are undetectable by standard summary metrics. Women using RT-CGM were able to achieve better daytime glucose control, reducing fetal exposure to maternal glucose.
Collapse
Affiliation(s)
- Eleanor M Scott
- Department of Clinical and Population Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, U.K.
| | - Denice S Feig
- Department of Medicine, Sinai Health System, Toronto, Ontario, Canada
| | - Helen R Murphy
- Division of Maternal Health, St Thomas' Hospital, King's College London, London, U.K
| | | | | |
Collapse
|
14
|
Crew RC, Mark PJ, Waddell BJ. Obesity Disrupts Rhythmic Clock Gene Expression in Maternal Adipose Tissue during Rat Pregnancy. J Biol Rhythms 2019; 33:289-301. [PMID: 29761750 DOI: 10.1177/0748730418772499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Obesity during pregnancy causes numerous maternal and fetal health complications, but the underlying mechanisms remain unclear. Adipose tissue dysfunction in obesity has previously been linked to disruption of the intrinsic adipose clock gene network that is crucial for normal metabolic function. This adipose clock also undergoes major change as part of the maternal metabolic adaptation to pregnancy, but whether this is affected by maternal obesity is unknown. Consequently, in this study we tested the hypothesis that obesity disturbs rhythmic gene expression in maternal adipose tissue across pregnancy. A rat model of maternal obesity was established by cafeteria (CAF) feeding, and adipose expression of clock genes and associated nuclear receptors ( Ppars and Pgc1α) was measured across days 15-16 and 21-22 of gestation (term = 23 days). CAF feeding suppressed the mesor and/or amplitude of adipose tissue clock genes (most notably Bmal1, Per2, and Rev-erbα) relative to chow-fed controls (CON) across both days of gestation. On day 15, the CAF diet also induced adipose Pparα, Pparδ, and Pgc1α rhythmicity but repressed that of Pparγ, while expression of Pparα, Pparδ, and Pgc1α was reduced at select time points. CAF mothers were hyperleptinemic at both stages of gestation, and at day 21 this effect was time-of-day dependent. Fetal plasma leptin exhibited clear rhythmicity, albeit with low amplitude, but interestingly these levels were unaffected by CAF feeding. Our data show that maternal obesity disrupts rhythmic expression of clock and metabolic genes in maternal adipose tissue and leads to maternal but not fetal hyperleptinemia.
Collapse
Affiliation(s)
- Rachael C Crew
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter J Mark
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Brendan J Waddell
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
15
|
Sun Q, Zhang G, Chen R, Li R, Wang H, Jiang A, Li Z, Kong L, Fonken LK, Rajagopalan S, Sun Q, Liu C. Central IKK2 Inhibition Ameliorates Air Pollution-Mediated Hepatic Glucose and Lipid Metabolism Dysfunction in Mice With Type II Diabetes. Toxicol Sci 2018; 164:240-249. [PMID: 29635361 PMCID: PMC6016715 DOI: 10.1093/toxsci/kfy079] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Previous studies supported a role of hypothalamic inflammation in fine ambient particulate matter (PM2.5) exposure-mediated diabetes development. We therefore investigated the effects of PM2.5 exposure on insulin resistance and the disorders of hepatic glucose and lipid metabolism via hypothalamic inflammation. KKAy mice, a genetically susceptible model of type II diabetes mellitus, were administered intra-cerebroventricularly with IKK2 inhibitor (IMD-0354) and were exposed to either concentrated PM2.5 or filtered air (FA) for 4 weeks simultaneously via a versatile aerosol concentration exposure system. At the end of the exposure, fasting blood glucose and serum insulin were evaluated before epididymal adipose tissue and liver were collected, flow cytometry, quantitative PCR and Western blot were performed at euthanasia. We observed that intracerebroventricular administration of IMD-0354 attenuated insulin resistance, inhibited macrophage polarization to M1 phenotype in epididymal adipose tissue in response to PM2.5 exposure. Although the treatment did not affect hepatic inflammation or endoplasmic reticulum stress, it inhibited the expression of the enzymes for gluconeogenesis and lipogenesis in the liver. Therefore, our current finding indicates an important role of hypothalamic inflammation in PM2.5 exposure-mediated hepatic glucose and lipid metabolism disorder.
Collapse
Affiliation(s)
- Qing Sun
- College of Public Health, Dalian Medical University, Dalian 116044, China
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guoqing Zhang
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Peoples’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Rucheng Chen
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ran Li
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huanhuan Wang
- Department of Basic Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Apei Jiang
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhenwei Li
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liya Kong
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Laura K Fonken
- Psychology and Neuroscience, Unviersity of Colorado Boulder, Boulder, Colorado 80309
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio 44106
| | - Qinghua Sun
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Division of Environmental Health Sciences, The Ohio State University, Columbus, Ohio 43210
| | - Cuiqing Liu
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Shanghai Key Laboratory of Meteorology and Health, Shanghai 200135, China
| |
Collapse
|
16
|
Gliniak CM, Brown JM, Noy N. The retinol-binding protein receptor STRA6 regulates diurnal insulin responses. J Biol Chem 2017; 292:15080-15093. [PMID: 28733465 DOI: 10.1074/jbc.m117.782334] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/17/2017] [Indexed: 01/06/2023] Open
Abstract
It has long been appreciated that insulin action is closely tied to circadian rhythms. However, the mechanisms that dictate diurnal insulin sensitivity in metabolic tissues are not well understood. Retinol-binding protein 4 (RBP4) has been implicated as a driver of insulin resistance in rodents and humans, and it has become an attractive drug target in type II diabetes. RBP4 is synthesized primarily in the liver where it binds retinol and transports it to tissues throughout the body. The retinol-RBP4 complex (holo-RBP) can be recognized by a cell-surface receptor known as stimulated by retinoic acid 6 (STRA6), which transports retinol into cells. Coupled to retinol transport, holo-RBP can activate STRA6-driven Janus kinase (JAK) signaling and downstream induction of signal transducer and activator of transcription (STAT) target genes. STRA6 signaling in white adipose tissue has been shown to inhibit insulin receptor responses. Here, we examined diurnal rhythmicity of the RBP4/STRA6 signaling axis and investigated whether STRA6 is necessary for diurnal variations in insulin sensitivity. We show that adipose tissue STRA6 undergoes circadian patterning driven in part by the nuclear transcription factor REV-ERBα. Furthermore, STRA6 is necessary for diurnal rhythmicity of insulin action and JAK/STAT signaling in adipose tissue. These findings establish that holo-RBP and its receptor STRA6 are potent regulators of diurnal insulin responses and suggest that the holo-RBP/STRA6 signaling axis may represent a novel therapeutic target in type II diabetes.
Collapse
Affiliation(s)
- Christy M Gliniak
- From the Department of Cellular and Molecular Medicine and.,the Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106
| | - J Mark Brown
- From the Department of Cellular and Molecular Medicine and .,the Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106.,the Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, Ohio 44195 and
| | - Noa Noy
- From the Department of Cellular and Molecular Medicine and
| |
Collapse
|
17
|
Kim S, Lee HS, Park HK, Linton JA, Lee JW, Lee H. Visceral adiposity and expression of clock genes in peripheral blood mononuclear cells: A pilot study. Chronobiol Int 2017. [PMID: 28650669 DOI: 10.1080/07420528.2017.1337780] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Increasing evidence suggests a close interrelationship between disrupted circadian rhythms and obesity and metabolic disturbances. In particular, abdominal obesity, which contributes to the pathogenesis of metabolic disease, is associated with disrupted clock gene expression. However, little is known about the relationship between clock gene expression and accurate computed tomography (CT)-based measurements of visceral adiposity. Therefore, we examined the relationship between expression of clock genes in peripheral blood mononuclear cells (PBMCs) with visceral and subcutaneous adiposity in 75 healthy overweight or obese individuals. PBMCs were obtained from blood samples collected at 8 AM, and gene expression was analyzed by real-time reverse transcription polymerase chain reaction. Visceral and subcutaneous adiposity were measured by CT. Our results showed that visceral fat area was significantly positively correlated with BMAL1 and CRY1 mRNA levels and significantly negatively correlated with CLOCK, PER2, PER3 and CRY2 mRNA levels. In contrast, subcutaneous fat area was not correlated with the expression of any of the clock genes analyzed. After adjusting for multiple variables, visceral fat area was significantly associated with the expression of BMAL1, PER2 and CRY1. Taken together, our results indicate that visceral adiposity, but not subcutaneous adiposity, correlates with expression of clock genes in PBMCs.
Collapse
Affiliation(s)
- Sue Kim
- a International Health Care Center, Severance Hospital , Yonsei University Health System , Seoul , Korea
| | - Hye-Sun Lee
- b Biostatistics Collaboration Unit , Yonsei University College of Medicine , Gangnam-gu , Seoul , Korea
| | - Hyun-Ki Park
- c Department of Clinical Nursing , Biobehavioral Research Center, Mo-Im Kim Nursing Research Institute, Yonsei University , Seoul , Korea
| | - John A Linton
- a International Health Care Center, Severance Hospital , Yonsei University Health System , Seoul , Korea.,d Department of Family Medicine , Severance Hospital, Yonsei University College of Medicine , Seoul , Korea
| | - Ji-Won Lee
- e Department of Family Medicine , Gangnam Severance Hospital, Yonsei University College of Medicine , Seoul , Korea
| | - Hyangkyu Lee
- c Department of Clinical Nursing , Biobehavioral Research Center, Mo-Im Kim Nursing Research Institute, Yonsei University , Seoul , Korea.,f Department of Clinical Nursing , Yonsei University College of Nursing , Seoul , Korea
| |
Collapse
|
18
|
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronizes these with daily cycles, feeding patterns also regulates circadian clocks. The clock genes and adipocytokines show circadian rhythmicity. Dysfunction of these genes are involved in the alteration of these adipokines during the development of obesity. Food availability promotes the stimuli associated with food intake which is a circadian oscillator outside of the suprachiasmatic nucleus (SCN). Its circadian rhythm is arranged with the predictable daily mealtimes. Food anticipatory activity is mediated by a self-sustained circadian timing and its principal component is food entrained oscillator. However, the hypothalamus has a crucial role in the regulation of energy balance rather than food intake. Fatty acids or their metabolites can modulate neuronal activity by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. The timing of three-meal schedules indicates close association with the plasma levels of insulin and preceding food availability. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition can lead to uncoupling of peripheral clocks from the central pacemaker and to the development of metabolic disorders. Metabolic dysfunction is associated with circadian disturbances at both central and peripheral levels and, eventual disruption of circadian clock functioning can lead to obesity. While CLOCK expression levels are increased with high fat diet-induced obesity, peroxisome proliferator-activated receptor (PPAR) alpha increases the transcriptional level of brain and muscle ARNT-like 1 (BMAL1) in obese subjects. Consequently, disruption of clock genes results in dyslipidemia, insulin resistance and obesity. Modifying the time of feeding alone can greatly affect body weight. Changes in the circadian clock are associated with temporal alterations in feeding behavior and increased weight gain. Thus, shift work is associated with increased risk for obesity, diabetes and cardio-vascular diseases as a result of unusual eating time and disruption of circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
19
|
Targeting of the circadian clock via CK1δ/ε to improve glucose homeostasis in obesity. Sci Rep 2016; 6:29983. [PMID: 27439882 PMCID: PMC4954991 DOI: 10.1038/srep29983] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/24/2016] [Indexed: 12/21/2022] Open
Abstract
Growing evidence indicates that disruption of our internal timing system contributes to the incidence and severity of metabolic diseases, including obesity and type 2 diabetes. This is perhaps not surprising since components of the circadian clockwork are tightly coupled to metabolic processes across the body. In the current study, we assessed the impact of obesity on the circadian system in mice at a behavioural and molecular level, and determined whether pharmacological targeting of casein kinase 1δ and ε (CK1δ/ε), key regulators of the circadian clock, can confer metabolic benefit. We demonstrate that although behavioural rhythmicity was maintained in diet-induced obesity (DIO), gene expression profiling revealed tissue-specific alteration to the phase and amplitude of the molecular clockwork. Clock function was most significantly attenuated in visceral white adipose tissue (WAT) of DIO mice, and was coincident with elevated tissue inflammation, and dysregulation of clock-coupled metabolic regulators PPARα/γ. Further, we show that daily administration of a CK1δ/ε inhibitor (PF-5006739) improved glucose tolerance in both DIO and genetic (ob/ob) models of obesity. These data further implicate circadian clock disruption in obesity and associated metabolic disturbance, and suggest that targeting of the clock represents a therapeutic avenue for the treatment of metabolic disorders.
Collapse
|
20
|
Honma K, Hikosaka M, Mochizuki K, Goda T. Loss of circadian rhythm of circulating insulin concentration induced by high-fat diet intake is associated with disrupted rhythmic expression of circadian clock genes in the liver. Metabolism 2016; 65:482-91. [PMID: 26975540 DOI: 10.1016/j.metabol.2015.12.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 11/19/2015] [Accepted: 12/12/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Peripheral clock genes show a circadian rhythm is correlated with the timing of feeding in peripheral tissues. It was reported that these clock genes are strongly regulated by insulin action and that a high-fat diet (HFD) intake in C57BL/6J mice for 21days induced insulin secretion during the dark phase and reduced the circadian rhythm of clock genes. In this study, we examined the circadian expression patterns of these clock genes in insulin-resistant animal models with excess secretion of insulin during the day. MATERIALS/METHODS We examined whether insulin resistance induced by a HFD intake for 80days altered blood parameters (glucose and insulin concentrations) and expression of mRNA and proteins encoded by clock and functional genes in the liver using male ICR mice. RESULTS Serum insulin concentrations were continuously higher during the day in mice fed a HFD than control mice. Expression of lipogenesis-related genes (Fas and Accβ) and the transcription factor Chrebp peaked at zeitgeber time (ZT)24 in the liver of control mice. A HFD intake reduced the expression of these genes at ZT24 and disrupted the circadian rhythm. Expression of Bmal1 and Clock, transcription factors that compose the core feedback loop, showed circadian variation and were synchronously associated with Fas gene expression in control mice, but not in those fed a HFD. CONCLUSIONS These results indicate that the disruption of the circadian rhythm of insulin secretion by HFD intake is closely associated with the disappearance of circadian expression of lipogenic and clock genes in the liver of mice.
Collapse
Affiliation(s)
- Kazue Honma
- Laboratory of Nutritional Physiology, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Maki Hikosaka
- Laboratory of Nutritional Physiology, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Kazuki Mochizuki
- Laboratory of Nutritional Physiology, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; Laboratory of Food and Nutritional Sciences, Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Toshinao Goda
- Laboratory of Nutritional Physiology, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan.
| |
Collapse
|
21
|
Shimizu I, Yoshida Y, Minamino T. A role for circadian clock in metabolic disease. Hypertens Res 2016; 39:483-91. [DOI: 10.1038/hr.2016.12] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 01/17/2016] [Accepted: 01/18/2016] [Indexed: 12/11/2022]
|
22
|
Zhang S, Zhao H, Ng MK. Functional Module Analysis for Gene Coexpression Networks with Network Integration. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2015; 12:1146-60. [PMID: 26451826 PMCID: PMC4664071 DOI: 10.1109/tcbb.2015.2396073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Network has been a general tool for studying the complex interactions between different genes, proteins, and other small molecules. Module as a fundamental property of many biological networks has been widely studied and many computational methods have been proposed to identify the modules in an individual network. However, in many cases, a single network is insufficient for module analysis due to the noise in the data or the tuning of parameters when building the biological network. The availability of a large amount of biological networks makes network integration study possible. By integrating such networks, more informative modules for some specific disease can be derived from the networks constructed from different tissues, and consistent factors for different diseases can be inferred. In this paper, we have developed an effective method for module identification from multiple networks under different conditions. The problem is formulated as an optimization model, which combines the module identification in each individual network and alignment of the modules from different networks together. An approximation algorithm based on eigenvector computation is proposed. Our method outperforms the existing methods, especially when the underlying modules in multiple networks are different in simulation studies. We also applied our method to two groups of gene coexpression networks for humans, which include one for three different cancers, and one for three tissues from the morbidly obese patients. We identified 13 modules with three complete subgraphs, and 11 modules with two complete subgraphs, respectively. The modules were validated through Gene Ontology enrichment and KEGG pathway enrichment analysis. We also showed that the main functions of most modules for the corresponding disease have been addressed by other researchers, which may provide the theoretical basis for further studying the modules experimentally.
Collapse
|
23
|
Scott EM. Circadian clocks, obesity and cardiometabolic function. Diabetes Obes Metab 2015; 17 Suppl 1:84-9. [PMID: 26332972 DOI: 10.1111/dom.12518] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/28/2015] [Indexed: 11/29/2022]
Abstract
Life on earth is governed by the continuous 24-h cycle of light and dark. Organisms have adapted to this environment with clear diurnal rhythms in their physiology and metabolism, enabling them to anticipate predictable environmental fluctuations over the day and to optimize the timing of relevant biological processes to this cycle. These rhythms are regulated by molecular circadian clocks, and current evidence suggests that interactions between the central and peripheral molecular clocks are important in metabolic and vascular functions. Disrupting this process through mutations in the core clock genes or by interfering with the environmental zeitgebers that entrain the clock appear to modulate the function of cells and tissues, leading to an increased risk for cardiometabolic disease.
Collapse
Affiliation(s)
- E M Scott
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine Clarendon Way, University of Leeds, Leeds, UK
| |
Collapse
|
24
|
Jakubowicz D, Wainstein J, Ahrén B, Bar-Dayan Y, Landau Z, Rabinovitz HR, Froy O. High-energy breakfast with low-energy dinner decreases overall daily hyperglycaemia in type 2 diabetic patients: a randomised clinical trial. Diabetologia 2015; 58:912-9. [PMID: 25724569 DOI: 10.1007/s00125-015-3524-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 01/28/2015] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS High-energy breakfast and reduced-energy dinner (Bdiet) significantly reduces postprandial glycaemia in obese non-diabetic individuals. Our objective was to test whether this meal schedule reduces postprandial hyperglycaemia (PPHG) in patients with type 2 diabetes by enhancing incretin and insulin levels when compared with high-energy dinner and reduced-energy breakfast (Ddiet). METHODS In a randomised, open label, crossover design performed in a clinic setting, 18 individuals (aged 30-70 years with BMI 22-35 kg/m(2)) with type 2 diabetes (<10 years duration) treated with metformin and/or diet were given either Bdiet or Ddiet for 7 days. Participants were randomised by a person not involved in the study using a coin flip. Postprandial levels of plasma glucose, insulin, C-peptide and intact and total glucagon-like peptide-1 (iGLP-1 and tGLP-1) were assessed. The Bdiet included 2,946 kJ breakfast, 2,523 kJ lunch and 858 kJ dinner. The Ddiet comprised 858 kJ breakfast, 2,523 kJ lunch and 2,946 kJ dinner. RESULTS Twenty-two individuals were randomised and 18 analysed. The AUC for glucose (AUCglucose) throughout the day was 20% lower, whereas AUCinsulin, AUCC-peptide and AUCtGLP-1 were 20% higher for the Bdiet than the Ddiet. Glucose AUC0-180min and its peak were both lower by 24%, whereas insulin AUC0-180min was 11% higher after the Bdiet than the Ddiet. This was accompanied by 30% higher tGLP-1 and 16% higher iGLP-1 levels. Despite the diets being isoenergetic, lunch resulted in lower glucose (by 21-25%) and higher insulin (by 23%) with the Bdiet vs Ddiet. CONCLUSIONS/INTERPRETATION High energy intake at breakfast is associated with significant reduction in overall PPHG in diabetic patients over the entire day. This dietary adjustment may have a therapeutic advantage for the achievement of optimal metabolic control and may have the potential for being preventive for cardiovascular and other complications of type 2 diabetes. Trial registration ClinicalTrials.gov NCT01977833 Funding No specific funding was received for the study.
Collapse
Affiliation(s)
- Daniela Jakubowicz
- Diabetes Unit, E. Wolfson Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Holon, 58100, Israel,
| | | | | | | | | | | | | |
Collapse
|
25
|
Chen L, Yang G. Recent advances in circadian rhythms in cardiovascular system. Front Pharmacol 2015; 6:71. [PMID: 25883568 PMCID: PMC4381645 DOI: 10.3389/fphar.2015.00071] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Growing evidence shows that intrinsic circadian clocks are tightly related to cardiovascular functions. The diurnal changes in blood pressure and heart rate are well known circadian rhythms. Endothelial function, platelet aggregation and thrombus formation exhibit circadian changes as well. The onset of many cardiovascular diseases (CVDs) or events, such as myocardial infarction, stroke, arrhythmia, and sudden cardiac death, also exhibits temporal trends. Furthermore, there is strong evidence from animal models and epidemiological studies showing that disruption of circadian rhythms is a significant risk factor for many CVDs, and the intervention of CVDs may have a time dependent effect. In this mini review, we summarized recent advances in our understanding of the relationship between circadian rhythm and cardiovascular physiology and diseases including blood pressure regulation and myocardial infarction.
Collapse
Affiliation(s)
- Lihong Chen
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Guangrui Yang
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
26
|
Ogai K, Matsumoto M, Minematsu T, Kitamura K, Kobayashi M, Sugama J, Sanada H. Development of an improved method for quantitative analysis of skin blotting: increasing reliability and applicability for skin assessment. Int J Cosmet Sci 2015; 37:425-32. [DOI: 10.1111/ics.12217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/07/2015] [Indexed: 01/08/2023]
Affiliation(s)
- K. Ogai
- Wellness Promotion Science Center; Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kanazawa Ishikawa Japan
| | - M. Matsumoto
- Division of Health Science; Department of Clinical Nursing; Graduate School of Medicine; Kanazawa University; Kanazawa Ishikawa Japan
| | - T. Minematsu
- Department of Gerontological Nursing/Wound Care Management; Graduate School of Medicine; The University of Tokyo; Bunkyo-ku Tokyo Japan
| | - K. Kitamura
- Department of Clinical Laboratory Science; Graduate School of Medical Science; Kanazawa University; Kanazawa Ishikawa Japan
| | - M. Kobayashi
- Wellness Promotion Science Center; Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kanazawa Ishikawa Japan
| | - J. Sugama
- Wellness Promotion Science Center; Institute of Medical, Pharmaceutical and Health Sciences; Kanazawa University; Kanazawa Ishikawa Japan
- Division of Health Science; Department of Clinical Nursing; Graduate School of Medicine; Kanazawa University; Kanazawa Ishikawa Japan
| | - H. Sanada
- Department of Gerontological Nursing/Wound Care Management; Graduate School of Medicine; The University of Tokyo; Bunkyo-ku Tokyo Japan
| |
Collapse
|
27
|
Sonnier T, Rood J, Gimble JM, Peterson CM. Glycemic control is impaired in the evening in prediabetes through multiple diurnal rhythms. J Diabetes Complications 2014; 28:836-43. [PMID: 24835190 DOI: 10.1016/j.jdiacomp.2014.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/11/2014] [Accepted: 04/01/2014] [Indexed: 01/12/2023]
Abstract
AIMS Recent studies suggest that circadian rhythms regulate glucose metabolism, weight loss, and even drug efficacy. Moreover, molecules targeted at the circadian clock show promise in treating metabolic disease. Therefore, this study set out to better characterize interactions among diurnal rhythms in prediabetes. METHODS Ten subjects with prediabetes completed oral glucose tolerance tests at 0700h and 1900h on the same day. Lipids and hormones were also measured. RESULTS Two-hour and three-hour glucose tolerances were worse in the evening by 40±52mg/dl (p=0.02) and 62±46mg/dl (p=0.001), respectively. These impairments were explained by lower insulin sensitivity (OGIS; 5.14±1.02 vs. 4.74±0.77mg/kg/min; p=0.03) and 2-hour AUC insulin levels (87.4±37.6 vs. 69.8±40.2mU∙hr/l; p=0.02) in the evening. Intriguingly, more insulin resistant subjects had weaker rhythms in insulin sensitivity (r=-0.66; p=0.04) but enhanced rhythms in insulin (r=-0.67; p=0.03) and cortisol (r=-0.78; p=0.008) levels. Importantly, the rhythms in cortisol primarily but also insulin sensitivity drove the declines in evening glucose tolerance (r=0.86; p=0.002). CONCLUSIONS Glycemic control is dramatically impaired in the evening in people with prediabetes, particularly when the cortisol rhythm is weak, but is unrelated to the rhythm in insulin levels. Therefore, food intake at dinnertime may need to be curbed in prediabetes.
Collapse
Affiliation(s)
- Tance Sonnier
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Jennifer Rood
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Jeffrey M Gimble
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA; Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA; LaCell LLC, New Orleans, LA 70112, USA
| | - Courtney M Peterson
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
28
|
Giovaninni NP, Fuly JT, Moraes LI, Coutinho TN, Trarbach EB, Jorge AADL, Costalonga EF. Study of the association between 3111T/C polymorphism of the CLOCK gene and the presence of overweight in schoolchildren. J Pediatr (Rio J) 2014; 90:500-5. [PMID: 24818524 DOI: 10.1016/j.jped.2014.01.011] [Citation(s) in RCA: 11] [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: 08/16/2013] [Revised: 11/20/2013] [Accepted: 01/21/2014] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES To evaluate the association between 3111T/C polymorphism of the CLOCK gene and the presence of obesity and sleep duration in children aged 6-13 years. In adults, this genetic variant has been associated with duration of sleep, ghrelin levels, weight, and eating habits. Although short sleep duration has been linked to obesity in children, no study has aimed to identify the possible molecular mechanisms of this association to date. METHODS Weight, height, and circumferences were transformed into Z-scores for age and gender. Genotyping was performed using TaqMan methodology. A questionnaire regarding hours of sleep was provided to parents. The appropriate statistical tests were performed. RESULTS This study evaluated 370 children (45% males, 55% females, mean age 8.5 ± 1.5 years). The prevalence of overweight was 18%. The duration of sleep was, on average, 9.7hours, and was inversely related to age (p<0.001). Genotype distribution was: 4% CC, 31% CT, and 65% TT. There was a trend toward higher prevalence of overweight in children who slept less than nine hours (23%) when compared to those who slept more than ten hours (16%, p=0.06). Genotype was not significantly correlated to any of the assessed outcomes. CONCLUSIONS The CLOCK 3111T/C polymorphism was not significantly associated with overweight or sleep duration in children in this city.
Collapse
Affiliation(s)
| | - Jeanne T Fuly
- Universidade Vila Velha (UVV), Vila Velha, ES, Brazil
| | | | | | | | | | | |
Collapse
|
29
|
Study of the association between 3111T/C polymorphism of the CLOCK gene and the presence of overweight in schoolchildren. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2014. [DOI: 10.1016/j.jpedp.2014.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
30
|
Abstract
In association with sleep-wake and fasting-feeding cycles, organisms experience dramatic oscillations in energetic demands and nutrient supply. It is therefore not surprising that various metabolic parameters, ranging from the activity status of molecular energy sensors to circulating nutrient levels, oscillate in time-of-day-dependent manners. It has become increasingly clear that rhythms in metabolic processes are not simply in response to daily environmental/behavioral influences, but are driven in part by cell autonomous circadian clocks. By synchronizing the cell with its environment, clocks modulate a host of metabolic processes in a temporally appropriate manner. The purpose of this article is to review current understanding of the interplay between circadian clocks and metabolism, in addition to the pathophysiologic consequences of disruption of this molecular mechanism, in terms of cardiometabolic disease development.
Collapse
Affiliation(s)
- Shannon M Bailey
- Division of Molecular and Cellular PathologyDepartment of PathologyDivision of Cardiovascular DiseasesDepartment of Medicine, University of Alabama at Birmingham, 703 19th Street South, ZRB 308, Birmingham, Alabama 35294, USA
| | - Uduak S Udoh
- Division of Molecular and Cellular PathologyDepartment of PathologyDivision of Cardiovascular DiseasesDepartment of Medicine, University of Alabama at Birmingham, 703 19th Street South, ZRB 308, Birmingham, Alabama 35294, USA
| | - Martin E Young
- Division of Molecular and Cellular PathologyDepartment of PathologyDivision of Cardiovascular DiseasesDepartment of Medicine, University of Alabama at Birmingham, 703 19th Street South, ZRB 308, Birmingham, Alabama 35294, USA
| |
Collapse
|
31
|
Wyse CA, Biello SM, Gill JMR. The bright-nights and dim-days of the urban photoperiod: implications for circadian rhythmicity, metabolism and obesity. Ann Med 2014; 46:253-63. [PMID: 24901354 DOI: 10.3109/07853890.2014.913422] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Artificial light decreases the amplitude of daily rhythms in human lifestyle principally by permitting activity and food intake to occur during hours of darkness, and allowing day-time activity to occur in dim light, indoors. Endogenous circadian timing mechanisms that oscillate with a period of 24 h have evolved to ensure physiology is synchronized with the daily variations in light, food, and social cues of the environment. Artificial light affects the synchronization between these oscillators, and metabolic disruption may be one consequence of this. By dampening the amplitude of environmental timing cues and disrupting circadian rhythmicity, artificial lighting might initiate metabolic disruption and contribute to the association between global urbanization and obesity. The aim of this review is to explore the historical, physiological, and epidemiological relationships between artificial light and circadian and metabolic dysfunction.
Collapse
Affiliation(s)
- Cathy A Wyse
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow , Glasgow G61 1QH , UK
| | | | | |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Accumulating evidence supports a role for the circadian clock in the development of metabolic disease. We discuss the influence of the circadian clock on glucose homeostasis, intermediary factors in this relationship, and potential therapies for the prevention or attenuation of metabolic disease associated with circadian misalignment. RECENT FINDINGS Murine studies with tissue-specific deletion of core clock genes in key metabolic tissues confirm a mechanistic relationship between the circadian clock and the development of metabolic disease. Circadian misalignment increases insulin resistance and decreases pancreatic function. Clock gene polymorphisms or altered expression of clock genes induced by circadian misalignment appear to play a role in the development of obesity and diabetes in humans. Circadian disruption caused by exposure to light at night is associated with lower nocturnal melatonin, which in turn seems to affect glucose metabolism. Potential therapies for circadian misalignment include entraining the central pacemaker with timed light exposure and/or melatonin and restricting food intake to the biological day. SUMMARY Completing the understanding of how genetic and environmental factors influence the circadian clock and the effect these have on human circadian metabolic physiology and disease will allow us to develop therapies for treating and preventing associated metabolic disease.
Collapse
Affiliation(s)
- Eberta Tan
- aDepartment of Diabetes and Endocrinology, Manny Cussins Centre, St James University Hospital, Beckett Wing bDivision of Cardiovascular and Diabetes Research, The Leeds Institute of Genetics Health and Therapeutics, Clarendon Way, University of Leeds, Leeds, UK
| | | |
Collapse
|
33
|
Portero McLellan KC, Wyne K, Villagomez ET, Hsueh WA. Therapeutic interventions to reduce the risk of progression from prediabetes to type 2 diabetes mellitus. Ther Clin Risk Manag 2014; 10:173-88. [PMID: 24672242 PMCID: PMC3964168 DOI: 10.2147/tcrm.s39564] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Clinical trials have demonstrated that it is possible to prevent diabetes through lifestyle modification, pharmacological intervention, and surgery. This review aims to summarize the effectiveness of these various therapeutic interventions in reducing the risk of progression of prediabetes to diabetes, and address the challenges to implement a diabetes prevention program at a community level. Strategies focusing on intensive lifestyle changes are not only efficient but cost-effective and/or cost-saving. Indeed, lifestyle intervention in people at high risk for type 2 diabetes mellitus (T2DM) has been successful in achieving sustained behavioral changes and a reduction in diabetes incidence even after the counseling is stopped. Although prediabetes is associated with health and economic burdens, it has not been adequately addressed by interventions or regulatory agencies in terms of prevention or disease management. Lifestyle intervention strategies to prevent T2DM should be distinct for different populations around the globe and should emphasize sex, age, ethnicity, and cultural and geographical considerations to be feasible and to promote better compliance. The translation of diabetes prevention research at a population level, especially finding the most effective methods of preventing T2DM in various societies and cultural settings remains challenging, but must be accomplished to stop this worldwide epidemic.
Collapse
Affiliation(s)
| | - Kathleen Wyne
- Division of Diabetes, Obesity and Lipids, Department of Medicine, The Methodist Hospital Diabetes and Metabolism Institute, and the Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, USA
| | - Evangelina Trejo Villagomez
- Division of Diabetes, Obesity and Lipids, Department of Medicine, The Methodist Hospital Diabetes and Metabolism Institute, and the Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, USA
| | - Willa A Hsueh
- Division of Diabetes, Obesity and Lipids, Department of Medicine, The Methodist Hospital Diabetes and Metabolism Institute, and the Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, USA
| |
Collapse
|
34
|
Maury E, Hong HK, Bass J. Circadian disruption in the pathogenesis of metabolic syndrome. DIABETES & METABOLISM 2014; 40:338-46. [PMID: 24433933 DOI: 10.1016/j.diabet.2013.12.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 12/15/2013] [Accepted: 12/16/2013] [Indexed: 12/19/2022]
Abstract
Metabolic syndrome is a multifactorial process induced by a combination of genetic and environmental factors and recent evidence has highlighted that circadian disruption and sleep loss contribute to disease pathogenesis. Emerging work in experimental genetic models has provided insight into the mechanistic basis for clock disruption in disease. Indeed, disruption of the clock system perturbs both neuroendocrine pathways within the hypothalamus important in feeding and energetics, in addition to peripheral tissues involved in glucose and lipid metabolism. This review illustrates the impact of molecular clock disruptions at the level of both brain and behavior and peripheral tissues, with a focus on how such dysregulation in turn impacts lipid and glucose homeostasis, inflammation and cardiovascular function. New insight into circadian biology may ultimately lead to improved therapeutics for metabolic syndrome and cardiovascular disease in humans.
Collapse
Affiliation(s)
- E Maury
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Lurie 7-220, Chicago, Illinois 60611, USA.
| | - H K Hong
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Lurie 7-220, Chicago, Illinois 60611, USA
| | - J Bass
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Lurie 7-220, Chicago, Illinois 60611, USA.
| |
Collapse
|
35
|
|
36
|
Naik A, Košir R, Rozman D. Genomic aspects of NAFLD pathogenesis. Genomics 2013; 102:84-95. [PMID: 23545492 DOI: 10.1016/j.ygeno.2013.03.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/19/2013] [Accepted: 03/22/2013] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most predominant liver disease worldwide and hepatic manifestation of the metabolic syndrome. Its histology spectrum ranges from steatosis, to steatohepatitis (NASH) that can further progress to cirrhosis and hepatocellular carcinoma (HCC). The increasing incidence of NAFLD has contributed to rising numbers of HCC occurrences. NAFLD progression is governed by genetic susceptibility, environmental factors, lifestyle and features of the metabolic syndrome, many of which overlap with HCC. Gene expression profiling and genome wide association studies have identified novel disease pathways and polymorphisms in genes that may be potential biomarkers of NAFLD progression. However, the multifactorial nature of NAFLD and the limited number of sufficiently powered studies are among the current limitations for validated biomarkers of clinical utility. Further studies incorporating the links between circadian regulation and hepatic metabolism might represent an additional direction in the search for predictive biomarkers of liver disease progression and treatment outcomes.
Collapse
Affiliation(s)
- Adviti Naik
- Faculty of Computer Sciences and Informatics, Tržaška Cesta 25, Ljubljana 1000, University of Ljubljana, Slovenia
| | | | | |
Collapse
|