1
|
Singh P, Gautam A, Trujillo M, Galligan J, Hensley L, Kapahi P, Bartke A. Growth Hormone Excess Drives Liver Aging via increased Glycation stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631635. [PMID: 39829894 PMCID: PMC11741365 DOI: 10.1101/2025.01.06.631635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Growth hormone (GH) plays a crucial role in various physiological functions, with its secretion tightly regulated by complex endocrine mechanisms. Pathological conditions such as acromegaly or pituitary tumors result in elevated circulating GH levels, which have been implicated in a spectrum of metabolic disorders, potentially by regulating liver metabolism. In this study, we focused on the liver, a key organ in metabolic regulation and a primary target of GH, to investigate the impact of high circulating GH on liver metabolism. We used bovine GH overexpressing transgenic (bGH-Tg) mice to conduct a comprehensive transcriptomic analysis of hepatic tissues. The bGH-Tg mouse livers exhibit dysregulated fatty acid metabolism and heightened inflammatory responses. Notably, the transcriptomic profile of young bGH-Tg mouse livers resembled that of aged livers and displayed markers of increased cellular senescence. Furthermore, these mice exhibited a significant accumulation of advanced glycation end products (AGEs). Intervention with glycation-lowering compounds effectively reversed the insulin resistance and aberrant transcriptomic signatures in the liver that are associated with elevated GH levels. These findings underscore the potential therapeutic value of glycation-lowering agents in mitigating the deleterious effects of chronic GH overexpression. Highlights Overexpression of bovine growth hormone impacts transcriptional changes in liver fat metabolism and inflammatory response in mice.High circulating growth hormone leads to transcriptional changes that suggest enhanced liver aging and induce cellular senescence.Detoxification pathways in bGH-Tg mice are inhibited, leading to the accumulation of Advanced Glycation End (AGE) products.Glycation-lowering compounds can mitigate pathologies associated with high GH levels.
Collapse
|
2
|
Jia X, Liu J, Jiang W, Chang L, Shen X, Jiang G, Li X, Chi C, Liu W, Zhang D. Binding site redundancy is critical for the regulation of fas by miR-30c in blunt snout bream (Megalobrama amblycephala). Comp Biochem Physiol A Mol Integr Physiol 2025; 299:111763. [PMID: 39395751 DOI: 10.1016/j.cbpa.2024.111763] [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: 09/11/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
MiR-30c and fatty acid synthase (fas) both play important roles in physiological processes such as lipid synthesis and fat metabolism. Predictive analysis revealed that fas is a target gene of miR-30c with multiple seed sites. Seed sites are useful to predict miRNA targeting relationships; however, detailed analyses of seed sites in fish genomes remain poorly studied. In this study, the regulatory relationship between miR-30c and fas, number and effect of seed regions, and mechanism by which miR-30c regulates lipid metabolism were evaluated in blunt snout bream (Megalobrama amblycephala). Four miR-30c target sites for fas were identified using various prediction tools. miR-30c mimics were transfected into 293 T cells, and dual-luciferase reporter assays were used to evaluate the roles of different fas target sites. When a single target site was mutated, relative luciferase activity was higher than that in the control group, with different activity levels depending on the mutation site. When multiple target sites were mutated, relative luciferase activity increased significantly as the number of mutation sites increased and was the highest when the four sites were mutated simultaneously. The miR-30c agomir was injected into the abdominal cavity of M. amblycephala at various concentrations for analyses of physiological and biochemical parameters in the liver and blood and the expression of genes related to lipid metabolism in the liver. Total cholesterol, free fatty acid, triglyceride, and low density lipoprotein levels were significantly lower after miR-30c agomir injection comparing to the control (P < 0.05). Additionally, the expression levels of genes related to lipid metabolism were significantly lower after miR-30c agomir injection than in the control (P < 0.05). In summary, this study identified four specific miR-30c target sites in the 3' UTR of fas mRNA; the effects of these sites are cumulative, and the redundancy ensures the accurate regulation of fas during evolution. In addition, miR-30c has a negative regulatory effect on fas and regulates lipid metabolism via various genes related to this process. Therefore, the regulation of miR-30c can effectively ameliorate the side effects of a high-fat diet on liver function in M. amblycephala.
Collapse
Affiliation(s)
- Xiaoyan Jia
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weibo Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Le Chang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxue Shen
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangzhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangfei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Cheng Chi
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenbin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dingdong Zhang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
3
|
Zheng ZJ, Zhang HY, Hu YL, Li Y, Wu ZH, Li ZP, Chen DR, Luo Y, Zhang XJ, Li C, Wang XY, Xu D, Qiu W, Li HP, Liao XP, Ren H, Sun J. Sleep Deprivation Induces Gut Damage via Ferroptosis. J Pineal Res 2024; 76:e12987. [PMID: 38975671 DOI: 10.1111/jpi.12987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Sleep deprivation (SD) has been associated with a plethora of severe pathophysiological syndromes, including gut damage, which recently has been elucidated as an outcome of the accumulation of reactive oxygen species (ROS). However, the spatiotemporal analysis conducted in this study has intriguingly shown that specific events cause harmful damage to the gut, particularly to goblet cells, before the accumulation of lethal ROS. Transcriptomic and metabolomic analyses have identified significant enrichment of metabolites related to ferroptosis in mice suffering from SD. Further analysis revealed that melatonin could rescue the ferroptotic damage in mice by suppressing lipid peroxidation associated with ALOX15 signaling. ALOX15 knockout protected the mice from the serious damage caused by SD-associated ferroptosis. These findings suggest that melatonin and ferroptosis could be targets to prevent devastating gut damage in animals exposed to SD. To sum up, this study is the first report that proposes a noncanonical modulation in SD-induced gut damage via ferroptosis with a clearly elucidated mechanism and highlights the active role of melatonin as a potential target to maximally sustain the state during SD.
Collapse
Affiliation(s)
- Zi-Jian Zheng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hai-Yi Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Ya-Lin Hu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yan Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Zhi-Hong Wu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Zhi-Peng Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dong-Rui Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Yang Luo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Cang Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Xiao-Yu Wang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Dan Xu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| |
Collapse
|
4
|
Lal H, Verma SK, Wang Y, Xie M, Young ME. Circadian Rhythms in Cardiovascular Metabolism. Circ Res 2024; 134:635-658. [PMID: 38484029 PMCID: PMC10947116 DOI: 10.1161/circresaha.123.323520] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 03/19/2024]
Abstract
Energetic demand and nutrient supply fluctuate as a function of time-of-day, in alignment with sleep-wake and fasting-feeding cycles. These daily rhythms are mirrored by 24-hour oscillations in numerous cardiovascular functional parameters, including blood pressure, heart rate, and myocardial contractility. It is, therefore, not surprising that metabolic processes also fluctuate over the course of the day, to ensure temporal needs for ATP, building blocks, and metabolism-based signaling molecules are met. What has become increasingly clear is that in addition to classic signal-response coupling (termed reactionary mechanisms), cardiovascular-relevant cells use autonomous circadian clocks to temporally orchestrate metabolic pathways in preparation for predicted stimuli/stresses (termed anticipatory mechanisms). Here, we review current knowledge regarding circadian regulation of metabolism, how metabolic rhythms are synchronized with cardiovascular function, and whether circadian misalignment/disruption of metabolic processes contribute toward the pathogenesis of cardiovascular disease.
Collapse
Affiliation(s)
- Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
5
|
Moreno-Cortés ML, Meza-Alvarado JE, García-Mena J, Hernández-Rodríguez A. Chronodisruption and Gut Microbiota: Triggering Glycemic Imbalance in People with Type 2 Diabetes. Nutrients 2024; 16:616. [PMID: 38474745 DOI: 10.3390/nu16050616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 03/14/2024] Open
Abstract
The desynchronization of physiological and behavioral mechanisms influences the gut microbiota and eating behavior in mammals, as shown in both rodents and humans, leading to the development of pathologies such as Type 2 diabetes (T2D), obesity, and metabolic syndrome. Recent studies propose resynchronization as a key input controlling metabolic cycles and contributing to reducing the risk of suffering some chronic diseases such as diabetes, obesity, or metabolic syndrome. In this analytical review, we present an overview of how desynchronization and its implications for the gut microbiome make people vulnerable to intestinal dysbiosis and consequent chronic diseases. In particular, we explore the eubiosis-dysbiosis phenomenon and, finally, propose some topics aimed at addressing chronotherapy as a key strategy in the prevention of chronic diseases.
Collapse
Affiliation(s)
- María Luisa Moreno-Cortés
- Laboratorio de Biomedicina, Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico
| | | | - Jaime García-Mena
- Departamento de Genética y Biología Molecular, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX 07360, Mexico
| | - Azucena Hernández-Rodríguez
- Laboratorio de Biomedicina, Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico
- Facultad de Bioanálisis, Universidad Veracruzana, Xalapa 91010, Veracruz, Mexico
| |
Collapse
|
6
|
Lee CH, Murrell CE, Chu A, Pan X. Circadian Regulation of Apolipoproteins in the Brain: Implications in Lipid Metabolism and Disease. Int J Mol Sci 2023; 24:17415. [PMID: 38139244 PMCID: PMC10743770 DOI: 10.3390/ijms242417415] [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: 11/02/2023] [Revised: 12/08/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
The circadian rhythm is a 24 h internal clock within the body that regulates various factors, including sleep, body temperature, and hormone secretion. Circadian rhythm disruption is an important risk factor for many diseases including neurodegenerative illnesses. The central and peripheral oscillators' circadian clock network controls the circadian rhythm in mammals. The clock genes govern the central clock in the suprachiasmatic nucleus (SCN) of the brain. One function of the circadian clock is regulating lipid metabolism. However, investigations of the circadian regulation of lipid metabolism-associated apolipoprotein genes in the brain are lacking. This review summarizes the rhythmic expression of clock genes and lipid metabolism-associated apolipoprotein genes within the SCN in Mus musculus. Nine of the twenty apolipoprotein genes identified from searching the published database (SCNseq and CircaDB) are highly expressed in the SCN. Most apolipoprotein genes (ApoE, ApoC1, apoA1, ApoH, ApoM, and Cln) show rhythmic expression in the brain in mice and thus might be regulated by the master clock. Therefore, this review summarizes studies on lipid-associated apolipoprotein genes in the SCN and other brain locations, to understand how apolipoproteins associated with perturbed cerebral lipid metabolism cause multiple brain diseases and disorders. This review describes recent advancements in research, explores current questions, and identifies directions for future research.
Collapse
Affiliation(s)
- Chaeeun Hannah Lee
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Charlotte Ellzabeth Murrell
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Alexander Chu
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Langone Hospital-Long Island, Mineola, NY 11501, USA
| |
Collapse
|
7
|
Khalifeh M, Santos RD, Oskuee RK, Badiee A, Aghaee-Bakhtiari SH, Sahebkar A. A novel regulatory facet for hypertriglyceridemia: The role of microRNAs in the regulation of triglyceride-rich lipoprotein biosynthesis. Prog Lipid Res 2023; 89:101197. [PMID: 36400247 DOI: 10.1016/j.plipres.2022.101197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the major leading global causes of death. Genetic and epidemiological studies strongly support the causal association between triacylglycerol-rich lipoproteins (TAGRL) and atherogenesis, even in statin-treated patients. Recent genetic evidence has clarified that variants in several key genes implicated in TAGRL metabolism are strongly linked to the increased ASCVD risk. There are several triacylglycerol-lowering agents; however, new therapeutic options are in development, among which are miRNA-based therapeutic approaches. MicroRNAs (miRNAs) are small non-coding RNAs (18-25 nucleotides) that negatively modulate gene expression through translational repression or degradation of target mRNAs, thereby reducing the levels of functional genes. MiRNAs play a crucial role in the development of hypertriglyceridemia as several miRNAs are dysregulated in both synthesis and clearance of TAGRL particles. MiRNA-based therapies in ASCVD have not yet been applied in human trials but are attractive. This review provides a concise overview of current interventions for hypertriglyceridemia and the development of novel miRNA and siRNA-based drugs. We summarize the miRNAs involved in the regulation of key genes in the TAGRLs synthesis pathway, which has gained attention as a novel target for therapeutic applications in CVD.
Collapse
Affiliation(s)
- Masoumeh Khalifeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raul D Santos
- Lipid Clinic Heart Institute (Incor), University of São Paulo, Medical School Hospital, São Paulo, Brazil
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Fawad JA, Luzader DH, Hanson GF, Moutinho TJ, McKinney CA, Mitchell PG, Brown-Steinke K, Kumar A, Park M, Lee S, Bolick DT, Medlock GL, Zhao JY, Rosselot AE, Chou CJ, Eshleman EM, Alenghat T, Hong CI, Papin JA, Moore SR. Histone Deacetylase Inhibition by Gut Microbe-Generated Short-Chain Fatty Acids Entrains Intestinal Epithelial Circadian Rhythms. Gastroenterology 2022; 163:1377-1390.e11. [PMID: 35934064 PMCID: PMC11551968 DOI: 10.1053/j.gastro.2022.07.051] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The circadian clock orchestrates ∼24-hour oscillations of gastrointestinal epithelial structure and function that drive diurnal rhythms in gut microbiota. Here, we use experimental and computational approaches in intestinal organoids to reveal reciprocal effects of gut microbial metabolites on epithelial timekeeping by an epigenetic mechanism. METHODS We cultured enteroids in media supplemented with sterile supernatants from the altered Schaedler Flora (ASF), a defined murine microbiota. Circadian oscillations of bioluminescent PER2 and Bmal1 were measured in the presence or absence of individual ASF supernatants. Separately, we applied machine learning to ASF metabolomics to identify phase-shifting metabolites. RESULTS Sterile filtrates from 3 of 7 ASF species (ASF360 Lactobacillus intestinalis, ASF361 Ligilactobacillus murinus, and ASF502 Clostridium species) induced minimal alterations in circadian rhythms, whereas filtrates from 4 ASF species (ASF356 Clostridium species, ASF492 Eubacterium plexicaudatum, ASF500 Pseudoflavonifactor species, and ASF519 Parabacteroides goldsteinii) induced profound, concentration-dependent phase shifts. Random forest classification identified short-chain fatty acid (SCFA) (butyrate, propionate, acetate, and isovalerate) production as a discriminating feature of ASF "shifters." Experiments with SCFAs confirmed machine learning predictions, with a median phase shift of 6.2 hours in murine enteroids. Pharmacologic or botanical histone deacetylase (HDAC) inhibitors yielded similar findings. Further, mithramycin A, an inhibitor of HDAC inhibition, reduced SCFA-induced phase shifts by 20% (P < .05) and conditional knockout of HDAC3 in enteroids abrogated butyrate effects on Per2 expression. Key findings were reproducible in human Bmal1-luciferase enteroids, colonoids, and Per2-luciferase Caco-2 cells. CONCLUSIONS Gut microbe-generated SCFAs entrain intestinal epithelial circadian rhythms by an HDACi-dependent mechanism, with critical implications for understanding microbial and circadian network regulation of intestinal epithelial homeostasis.
Collapse
Affiliation(s)
- Jibraan A Fawad
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Deborah H Luzader
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Gabriel F Hanson
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Thomas J Moutinho
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Craig A McKinney
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Paul G Mitchell
- University of Virginia School of Medicine, Charlottesville, Virginia
| | - Kathleen Brown-Steinke
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Ajay Kumar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Miri Park
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Suengwon Lee
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - David T Bolick
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Greg L Medlock
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | - Jesse Y Zhao
- University of Virginia School of Medicine, Charlottesville, Virginia
| | - Andrew E Rosselot
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - C James Chou
- College of Pharmacy, Medical University of South Carolina, Charleston, South Carolina
| | - Emily M Eshleman
- Division of Immunobiology, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Theresa Alenghat
- Division of Immunobiology, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Christian I Hong
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Sean R Moore
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
9
|
Vagus nerve stimulation increases stomach-brain coupling via a vagal afferent pathway. Brain Stimul 2022; 15:1279-1289. [PMID: 36067977 DOI: 10.1016/j.brs.2022.08.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Maintaining energy homeostasis is vital and supported by vagal signaling between digestive organs and the brain. Previous research has established a gastric network in the brain that is phase synchronized with the rhythm of the stomach, but tools to perturb its function were lacking. OBJECTIVE To evaluate whether stomach-brain coupling can be acutely increased by non-invasively stimulating vagal afferent projections to the brain. METHODS Using a single-blind randomized crossover design, we investigated the effect of acute right-sided transcutaneous auricular vagus nerve stimulation (taVNS) versus sham stimulation on stomach-brain coupling. RESULTS In line with preclinical research, taVNS increased stomach-brain coupling in the nucleus of the solitary tract (NTS) and the midbrain while boosting coupling across the brain. Crucially, in the cortex, taVNS-induced changes in coupling occurred primarily in transmodal regions and were associated with changes in hunger ratings as indicators of the subjective metabolic state. CONCLUSIONS taVNS increases stomach-brain coupling via an NTS-midbrain pathway that signals gut-induced reward, indicating that communication between the brain and the body is effectively modulated by vago-vagal signaling. Such insights may help us better understand the role of vagal afferents in orchestrating the recruitment of the gastric network which could pave the way for novel neuromodulatory treatments.
Collapse
|
10
|
Peng L, Wen L, Zhang J, Zhang X, Wei Q, Guo J, Zeng J. Circadian Pharmacological Effects of Paeoniflorin on Mice With Urticaria-like Lesions. Front Pharmacol 2022; 12:639580. [PMID: 35222003 PMCID: PMC8863972 DOI: 10.3389/fphar.2021.639580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/22/2021] [Indexed: 12/29/2022] Open
Abstract
Paeoniflorin (PF) is a monoterpene glucoside with various biological properties, and it suppresses allergic and inflammatory responses in a rat model of urticaria-like lesions (UL). In the present study, we treated OVA-induced mice presenting UL with PF at four circadian time points (ZT22, ZT04, ZT10, and ZT16) to determine the optimal administration time of PF. The pharmacological effects of PF were assessed by analyzing the scratching behavior; histopathological features; allergic responses such as immunoglobulin E (IgE), leukotriene B4 (LTB4), and histamine (HIS) release; inflammatory cell infiltration [mast cell tryptase (MCT) and eosinophil protein X (EPX)]; and mRNA levels of inflammatory cytokines such as interleukin (IL)-12, IL-6, interferon-γ (IFN-γ), and IL-4. It was demonstrated that PF significantly alleviated scratching behavior and histopathological features, and ZT10 dosing was the most effective time point in remission of the condition among the four circadian time points. Moreover, PF decreased the serum levels of IgE, LTB4, and HIS, and PF administration at ZT10 produced relatively superior effectiveness. PF treatment, especially dosing at ZT10, significantly reduced the number of mast cells and granules and diminished the infiltration of MCT and EPX in the skin tissues of mice with UL. Furthermore, the oral administration of PF effectively decreased the inflammatory cytokine levels of IL-12 mRNA. In conclusion, different administration times of PF affected its efficacy in mice with UL. ZT10 administration demonstrated relatively superior effectiveness, and it might be the optimal administration time for the treatment of urticaria.
Collapse
Affiliation(s)
- Li Peng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijuan Wen
- Clinical Skills Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaotong Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin Wei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Guo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Dermatological Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jing Guo, ; Jinhao Zeng,
| | - Jinhao Zeng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Geriatric Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Jing Guo, ; Jinhao Zeng,
| |
Collapse
|
11
|
Pan X. Cholesterol Metabolism in Chronic Kidney Disease: Physiology, Pathologic Mechanisms, and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:119-143. [PMID: 35503178 PMCID: PMC11106795 DOI: 10.1007/978-981-19-0394-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
12
|
Allaband C, Lingaraju A, Martino C, Russell B, Tripathi A, Poulsen O, Dantas Machado AC, Zhou D, Xue J, Elijah E, Malhotra A, Dorrestein PC, Knight R, Haddad GG, Zarrinpar A. Intermittent Hypoxia and Hypercapnia Alter Diurnal Rhythms of Luminal Gut Microbiome and Metabolome. mSystems 2021; 6:e0011621. [PMID: 34184915 PMCID: PMC8269208 DOI: 10.1128/msystems.00116-21] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Obstructive sleep apnea (OSA), characterized by intermittent hypoxia and hypercapnia (IHC), affects the composition of the gut microbiome and metabolome. The gut microbiome has diurnal oscillations that play a crucial role in regulating circadian and overall metabolic homeostasis. Thus, we hypothesized that IHC adversely alters the gut luminal dynamics of key microbial families and metabolites. The objective of this study was to determine the diurnal dynamics of the fecal microbiome and metabolome of Apoe-/- mice after a week of IHC exposure. Individually housed, 10-week-old Apoe-/- mice on an atherogenic diet were split into two groups. One group was exposed to daily IHC conditions for 10 h (Zeitgeber time 2 [ZT2] to ZT12), while the other was maintained in room air. Six days after the initiation of the IHC conditions, fecal samples were collected every 4 h for 24 h (6 time points). We performed 16S rRNA gene amplicon sequencing and untargeted liquid chromatography-mass spectrometry (LC-MS) to assess changes in the microbiome and metabolome. IHC induced global changes in the cyclical dynamics of the gut microbiome and metabolome. Ruminococcaceae, Lachnospiraceae, S24-7, and Verrucomicrobiaceae had the greatest shifts in their diurnal oscillations. In the metabolome, bile acids, glycerolipids (phosphocholines and phosphoethanolamines), and acylcarnitines were greatly affected. Multi-omic analysis of these results demonstrated that Ruminococcaceae and tauro-β-muricholic acid (TβMCA) cooccur and are associated with IHC conditions and that Coriobacteriaceae and chenodeoxycholic acid (CDCA) cooccur and are associated with control conditions. IHC significantly change the diurnal dynamics of the fecal microbiome and metabolome, increasing members and metabolites that are proinflammatory and proatherogenic while decreasing protective ones. IMPORTANCE People with obstructive sleep apnea are at a higher risk of high blood pressure, type 2 diabetes, cardiac arrhythmias, stroke, and sudden cardiac death. We wanted to understand whether the gut microbiome changes induced by obstructive sleep apnea could potentially explain some of these medical problems. By collecting stool from a mouse model of this disease at multiple time points during the day, we studied how obstructive sleep apnea changed the day-night patterns of microbes and metabolites of the gut. Since the oscillations of the gut microbiome play a crucial role in regulating metabolism, changes in these oscillations can explain why these patients can develop so many metabolic problems. We found changes in microbial families and metabolites that regulate many metabolic pathways contributing to the increased risk for heart disease seen in patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Celeste Allaband
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Amulya Lingaraju
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
| | - Cameron Martino
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Baylee Russell
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
| | - Anupriya Tripathi
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Orit Poulsen
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | | | - Dan Zhou
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Jin Xue
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Emmanuel Elijah
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Atul Malhotra
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
| | - Pieter C. Dorrestein
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Rob Knight
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California, USA
| | - Gabriel G. Haddad
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Department of Neuroscience, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, California, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
- VA Health Sciences San Diego, La Jolla, California, USA
| |
Collapse
|
13
|
Parnell AA, De Nobrega AK, Lyons LC. Translating around the clock: Multi-level regulation of post-transcriptional processes by the circadian clock. Cell Signal 2021; 80:109904. [PMID: 33370580 PMCID: PMC8054296 DOI: 10.1016/j.cellsig.2020.109904] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022]
Abstract
The endogenous circadian clock functions to maintain optimal physiological health through the tissue specific coordination of gene expression and synchronization between tissues of metabolic processes throughout the 24 hour day. Individuals face numerous challenges to circadian function on a daily basis resulting in significant incidences of circadian disorders in the United States and worldwide. Dysfunction of the circadian clock has been implicated in numerous diseases including cancer, diabetes, obesity, cardiovascular and hepatic abnormalities, mood disorders and neurodegenerative diseases. The circadian clock regulates molecular, metabolic and physiological processes through rhythmic gene expression via transcriptional and post-transcriptional processes. Mounting evidence indicates that post-transcriptional regulation by the circadian clock plays a crucial role in maintaining tissue specific biological rhythms. Circadian regulation affecting RNA stability and localization through RNA processing, mRNA degradation, and RNA availability for translation can result in rhythmic protein synthesis, even when the mRNA transcripts themselves do not exhibit rhythms in abundance. The circadian clock also targets the initiation and elongation steps of translation through multiple pathways. In this review, the influence of the circadian clock across the levels of post-transcriptional, translation, and post-translational modifications are examined using examples from humans to cyanobacteria demonstrating the phylogenetic conservation of circadian regulation. Lastly, we briefly discuss chronotherapies and pharmacological treatments that target circadian function. Understanding the complexity and levels through which the circadian clock regulates molecular and physiological processes is important for future advancement of therapeutic outcomes.
Collapse
Affiliation(s)
- Amber A Parnell
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Aliza K De Nobrega
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
14
|
Pan X, Taylor MJ, Cohen E, Hanna N, Mota S. Circadian Clock, Time-Restricted Feeding and Reproduction. Int J Mol Sci 2020; 21:ijms21030831. [PMID: 32012883 PMCID: PMC7038040 DOI: 10.3390/ijms21030831] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022] Open
Abstract
The goal of this review was to seek a better understanding of the function and differential expression of circadian clock genes during the reproductive process. Through a discussion of how the circadian clock is involved in these steps, the identification of new clinical targets for sleep disorder-related diseases, such as reproductive failure, will be elucidated. Here, we focus on recent research findings regarding circadian clock regulation within the reproductive system, shedding new light on circadian rhythm-related problems in women. Discussions on the roles that circadian clock plays in these reproductive processes will help identify new clinical targets for such sleep disorder-related diseases.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
- Correspondence:
| | - Meredith J. Taylor
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Emma Cohen
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Nazeeh Hanna
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Department of Pediatrics, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York, NY 11501, USA
- Diabetes and Obesity Research Center, NYU Winthrop Hospital, Mineola, New York, NY 11501, USA
| |
Collapse
|
15
|
Iqbal J, Jahangir Z, Al-Qarni AA. Microsomal Triglyceride Transfer Protein: From Lipid Metabolism to Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:37-52. [DOI: 10.1007/978-981-15-6082-8_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Pan X, Mota S, Zhang B. Circadian Clock Regulation on Lipid Metabolism and Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:53-66. [PMID: 32705594 PMCID: PMC8593891 DOI: 10.1007/978-981-15-6082-8_5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The basic helix-loop-helix-PAS transcription factor (CLOCK, Circadian locomotor output cycles protein kaput) was discovered in 1994 as a circadian clock. Soon after its discovery, the circadian clock, Aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL, also call BMAL1), was shown to regulate adiposity and body weight by controlling on the brain hypothalamic suprachiasmatic nucleus (SCN). Farther, circadian clock genes were determined to exert several of lipid metabolic and diabetes effects, overall indicating that CLOCK and BMAL1 act as a central master circadian clock. A master circadian clock acts through the neurons and hormones, with expression in the intestine, liver, kidney, lung, heart, SCN of brain, and other various cell types of the organization. Among circadian clock genes, numerous metabolic syndromes are the most important in the regulation of food intake (via regulation of circadian clock genes or clock-controlled genes in peripheral tissue), which lead to a variation in plasma phospholipids and tissue phospholipids. Circadian clock genes affect the regulation of transporters and proteins included in the regulation of phospholipid metabolism. These genes have recently received increasing recognition because a pharmacological target of circadian clock genes may be of therapeutic worth to make better resistance against insulin, diabetes, obesity, metabolism syndrome, atherosclerosis, and brain diseases. In this book chapter, we focus on the regulation of circadian clock and summarize its phospholipid effect as well as discuss the chemical, physiology, and molecular value of circadian clock pathway regulation for the treatment of plasma lipids and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA.
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA
| | - Boyang Zhang
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA
| |
Collapse
|
17
|
Qin C, Sun J, Wang J, Han Y, Yang H, Shi Q, Lv Y, Hu P. Discovery of differentially expressed genes in the intestines of Pelteobagrus vachellii within a light/dark cycle. Chronobiol Int 2019; 37:339-352. [PMID: 31809585 DOI: 10.1080/07420528.2019.1690498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In aquaculture, it is necessary to determine of the diurnal biological variations in the intestines to determine an appropriate feeding schedule. The present study aimed to examine the transcriptomes of the Pelteobagrus vachellii intestines at four time points (0 h, 6 h, 12 h, and 18 h) within a light/dark cycle. In comparison with the zeitgeber time 0 (ZT0) transcriptomes, we identified 37,842 unigenes with significant differential expression, including 6,638; 9,626; and 7,938 that genes upregulated, and 3,507; 4,703; and 5,412 genes that were down regulated at 4, 12, and 24 h respectively. The differentially expressed unigenes were subjected to enrichment analysis, which indicated the involvement of the major digestive pathways, including digestion of protein, lipid and carbohydrate, catabolic process (protein, carbohydrate and lipid), and circadian rhythm. We selected 73 key differentially expressed genes (DEGs) from among these pathways and identified DEGs that showed increased expression at night, including those encoding trypsin-3, chymotrypsinogen 2, amino acid transporter, maltase-glucoamylase, facilitated glucose transporter, lipase, phospholipase, fatty acid-binding protein, fatty acid synthase, long-chain fatty acid transport protein, and apolipoprotein. Moreover, DEGs involved of circadian rhythm were identified, including brain-muscle-Arnt-like 1 (BMAL1), cryptochrome-1, circadian locomoter output cycles protein kaput (CLOCK) and period circadian protein homolog 1-3. Finally, the expression levels of 12 unigenes were analyzed using quantitative real-time PCR, which were in accordance with RNA-sequencing analysis. In general, the expression of genes related to the digestion of proteins, lipids, and carbohydrates showed upregulated expression at night; however, the peak time of expression of transporters for different nutrition molecules showed more diversification within the light/dark cycle.
Collapse
Affiliation(s)
- Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | | | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | | | - He Yang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | - Qingchao Shi
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | - Yunyun Lv
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| | - Peng Hu
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang, PR China.,College of Life Science, Neijiang Normal University, Neijiang, PR China
| |
Collapse
|
18
|
Sussman W, Stevenson M, Mowdawalla C, Mota S, Ragolia L, Pan X. BMAL1 controls glucose uptake through paired-homeodomain transcription factor 4 in differentiated Caco-2 cells. Am J Physiol Cell Physiol 2019; 317:C492-C501. [PMID: 31216190 PMCID: PMC6766619 DOI: 10.1152/ajpcell.00058.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023]
Abstract
The transcription factor aryl hydrocarbon receptor nuclear translocator-like protein-1 (BMAL1) is an essential regulator of the circadian clock, which controls the 24-h cycle of physiological processes such as nutrient absorption. To examine the role of BMAL1 in small intestinal glucose absorption, we used differentiated human colon adenocarcinoma cells (Caco-2 cells). Here, we show that BMAL1 regulates glucose uptake in differentiated Caco-2 cells and that this process is dependent on the glucose transporter sodium-glucose cotransporter 1 (SGLT1). Mechanistic studies show that BMAL1 regulates glucose uptake by controlling the transcription of SGLT1 involving paired-homeodomain transcription factor 4 (PAX4), a transcriptional repressor. This is supported by the observation that clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated endonuclease Cas9 (Cas9) knockdown of PAX4 increases SGLT1 and glucose uptake. Chromatin immunoprecipitation (ChIP) and ChIP-quantitative PCR assays show that the knockdown or overexpression of BMAL1 decreases or increases the binding of PAX4 to the hepatocyte nuclear factor 1-α binding site of the SGLT1 promoter, respectively. These findings identify BMAL1 as a critical mediator of small intestine carbohydrate absorption and SGLT1.
Collapse
Affiliation(s)
- Whitney Sussman
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Matthew Stevenson
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Cyrus Mowdawalla
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Louis Ragolia
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
19
|
Pan X, Schwartz GJ, Hussain MM. Oleoylethanolamide differentially regulates glycerolipid synthesis and lipoprotein secretion in intestine and liver. J Lipid Res 2018; 59:2349-2359. [PMID: 30369486 DOI: 10.1194/jlr.m089250] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/26/2018] [Indexed: 01/13/2023] Open
Abstract
Dietary fat absorption takes place in the intestine, and the liver mobilizes endogenous fat to other tissues by synthesizing lipoproteins that require apoB and microsomal triglyceride transfer protein (MTP). Dietary fat triggers the synthesis of oleoylethanolamide (OEA), a regulatory fatty acid that signals satiety to reduce food intake mainly by enhancing neural PPARα activity, in enterocytes. We explored OEA's roles in the assembly of lipoproteins in WT and Ppara -/- mouse enterocytes and hepatocytes, Caco-2 cells, and human liver-derived cells. In differentiated Caco-2 cells, OEA increased synthesis and secretion of triacylglycerols, apoB secretion in chylomicrons, and MTP expression in a dose-dependent manner. OEA also increased MTP activity and triacylglycerol secretion in WT and knockout primary enterocytes. In contrast to its intestinal cell effects, OEA reduced synthesis and secretion of triacylglycerols, apoB secretion, and MTP expression and activity in human hepatoma Huh-7 and HepG2 cells. Also, OEA reduced MTP expression and triacylglycerol secretion in WT, but not knockout, primary hepatocytes. These studies indicate differential effects of OEA on lipid synthesis and lipoprotein assembly: in enterocytes, OEA augments glycerolipid synthesis and lipoprotein assembly independent of PPARα. Conversely, in hepatocytes, OEA reduces MTP expression, glycerolipid synthesis, and lipoprotein secretion through PPARα-dependent mechanisms.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY .,Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY
| | - Gary J Schwartz
- Departments of Medicine and Neuroscience, Albert Einstein College of Medicine, Bronx, NY
| | - M Mahmood Hussain
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY .,Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY.,Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY
| |
Collapse
|
20
|
Fortes-Silva R, Valle SVD, Lopéz-Olmeda JF. Daily rhythms of swimming activity, synchronization to different feeding times and effects on anesthesia practice in an Amazon fish species (Colossoma macropomum). Chronobiol Int 2018; 35:1713-1722. [PMID: 30141973 DOI: 10.1080/07420528.2018.1509078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
This study aimed to investigate the existence of day-night differences in the time for anesthesia and recovery in tambaqui exposed to the anesthetic eugenol and the influence of feeding time. Thus, we evaluated: (1) swimming activity; (2) food anticipatory activity (FAA) as a synchronizer of swimming activity and change to susceptibility to anesthetic; and (3) the effects of diurnal/nocturnal anesthesia exposure of fish feeding in the mid-light phase: 12:00 h (ML) and fish feeding in the mid-dark phase: 00:00 h (MD). Our findings revealed strictly nocturnal activity for tambaqui (94.2%), known as diurnal fish to date. Moreover, FAA was observed in tambaqui fed at MD, which showed a sustained increase in activity that began 2 h before feeding time and lasted until feeding. In contrast, no FAA was observed in fish fed at ML. Regarding anesthesia by day or night, the tambaqui treated with eugenol exhibited no difference in induction time. However, differences were observed in recovery times, with fish anesthetized at day recovering in 1-2 min and fish anesthetized at night recovering in 5-7 min. In short, our findings revealed for the first time the nocturnal behavior of tambaqui. These results indicated that recovery by day/night by eugenol in tambaqui has a strong dependence of behavioral patterns and the time of day.
Collapse
Affiliation(s)
- Rodrigo Fortes-Silva
- a Laboratory of Feeding Behavior and Fish Nutrition (AquaUFRB), Faculty of Fish Engineering (NEPA), Center of Agricultural Sciences, Environmental and Biological(CCAAB) , Federal University of Bahia (UFRB) , Bahia , Brazil
| | - Silvan Vianna Do Valle
- a Laboratory of Feeding Behavior and Fish Nutrition (AquaUFRB), Faculty of Fish Engineering (NEPA), Center of Agricultural Sciences, Environmental and Biological(CCAAB) , Federal University of Bahia (UFRB) , Bahia , Brazil
| | - Jose Fernando Lopéz-Olmeda
- b Department of Physiology, Faculty of Biology, Regional Campus of International Excellence "Campus Mare Nostrum" , University of Murcia , Murcia , Spain
| |
Collapse
|
21
|
Duszka K, Wahli W. Enteric Microbiota⁻Gut⁻Brain Axis from the Perspective of Nuclear Receptors. Int J Mol Sci 2018; 19:ijms19082210. [PMID: 30060580 PMCID: PMC6121494 DOI: 10.3390/ijms19082210] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors (NRs) play a key role in regulating virtually all body functions, thus maintaining a healthy operating body with all its complex systems. Recently, gut microbiota emerged as major factor contributing to the health of the whole organism. Enteric bacteria have multiple ways to influence their host and several of them involve communication with the brain. Mounting evidence of cooperation between gut flora and NRs is already available. However, the full potential of the microbiota interconnection with NRs remains to be uncovered. Herewith, we present the current state of knowledge on the multifaceted roles of NRs in the enteric microbiota–gut–brain axis.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological, 11 Mandalay Road, Singapore 308232, Singapore.
- Center for Integrative Genomics, University of Lausanne, Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
22
|
de Assis LVM, Moraes MN, Magalhães-Marques KK, Kinker GS, da Silveira Cruz-Machado S, Castrucci AMDL. Non-Metastatic Cutaneous Melanoma Induces Chronodisruption in Central and Peripheral Circadian Clocks. Int J Mol Sci 2018; 19:E1065. [PMID: 29614021 PMCID: PMC5979525 DOI: 10.3390/ijms19041065] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 12/11/2022] Open
Abstract
The biological clock has received increasing interest due to its key role in regulating body homeostasis in a time-dependent manner. Cancer development and progression has been linked to a disrupted molecular clock; however, in melanoma, the role of the biological clock is largely unknown. We investigated the effects of the tumor on its micro- (TME) and macro-environments (TMaE) in a non-metastatic melanoma model. C57BL/6J mice were inoculated with murine B16-F10 melanoma cells and 2 weeks later the animals were euthanized every 6 h during 24 h. The presence of a localized tumor significantly impaired the biological clock of tumor-adjacent skin and affected the oscillatory expression of genes involved in light- and thermo-reception, proliferation, melanogenesis, and DNA repair. The expression of tumor molecular clock was significantly reduced compared to healthy skin but still displayed an oscillatory profile. We were able to cluster the affected genes using a human database and distinguish between primary melanoma and healthy skin. The molecular clocks of lungs and liver (common sites of metastasis), and the suprachiasmatic nucleus (SCN) were significantly affected by tumor presence, leading to chronodisruption in each organ. Taken altogether, the presence of non-metastatic melanoma significantly impairs the organism's biological clocks. We suggest that the clock alterations found in TME and TMaE could impact development, progression, and metastasis of melanoma; thus, making the molecular clock an interesting pharmacological target.
Collapse
Affiliation(s)
- Leonardo Vinícius Monteiro de Assis
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | - Maria Nathália Moraes
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | - Keila Karoline Magalhães-Marques
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | - Gabriela Sarti Kinker
- Laboratory of Chronopharmacology, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | - Sanseray da Silveira Cruz-Machado
- Laboratory of Chronopharmacology, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-900, Brazil.
| | - Ana Maria de Lauro Castrucci
- Laboratory of Comparative Physiology of Pigmentation, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-900, Brazil.
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
23
|
Glucocorticoids Drive Diurnal Oscillations in T Cell Distribution and Responses by Inducing Interleukin-7 Receptor and CXCR4. Immunity 2018; 48:286-298.e6. [PMID: 29396162 DOI: 10.1016/j.immuni.2018.01.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 09/05/2017] [Accepted: 12/29/2017] [Indexed: 12/23/2022]
Abstract
Glucocorticoids are steroid hormones with strong anti-inflammatory and immunosuppressive effects that are produced in a diurnal fashion. Although glucocorticoids have the potential to induce interleukin-7 receptor (IL-7R) expression in T cells, whether they control T cell homeostasis and responses at physiological concentrations remains unclear. We found that glucocorticoid receptor signaling induces IL-7R expression in mouse T cells by binding to an enhancer of the IL-7Rα locus, with a peak at midnight and a trough at midday. This diurnal induction of IL-7R supported the survival of T cells and their redistribution between lymph nodes, spleen, and blood by controlling expression of the chemokine receptor CXCR4. In mice, T cell accumulation in the spleen at night enhanced immune responses against soluble antigens and systemic bacterial infection. Our results reveal the immunoenhancing role of glucocorticoids in adaptive immunity and provide insight into how immune function is regulated by the diurnal rhythm.
Collapse
|
24
|
Sharbati J, Bohmer M, Bohmer N, Keller A, Backes C, Franke A, Steinberg P, Zeljenková D, Einspanier R. Transcriptomic Analysis of Intestinal Tissues from Two 90-Day Feeding Studies in Rats Using Genetically Modified MON810 Maize Varieties. Front Genet 2017; 8:222. [PMID: 29312443 PMCID: PMC5742243 DOI: 10.3389/fgene.2017.00222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022] Open
Abstract
Background: Global as well as specific expression profiles of selected rat tissues were characterized to assess the safety of genetically modified (GM) maize MON810 containing the insecticidal protein Cry1Ab. Gene expression was evaluated by use of Next Generation Sequencing (NGS) as well as RT-qPCR within rat intestinal tissues based on mandatory 90-day rodent feeding studies. In parallel to two 90-day feeding studies, the transcriptional response of rat tissues was assessed as another endpoint to enhance the mechanistic interpretation of GM feeding studies and/or to facilitate the generation of a targeted hypothesis. Rats received diets containing 33% GM maize (MON810) or near-isogenic control maize. As a site of massive exposure to ingested feed the transcriptomic response of ileal and colonic tissue was profiled via RT-qPCR arrays targeting apoptosis, DNA-damage/repair, unfolded protein response (UPR). For global RNA profiling of rat ileal tissue, we applied NGS. Results: No biological response to the GM-diet was observed in male and in female rat tissues. Transcriptome wide analysis of gene expression by RNA-seq confirmed these findings. Nevertheless, gene ontology (GO) analysis clearly associated a set of distinctly regulated transcripts with circadian rhythms. We confirmed differential expression of circadian clock genes using RT-qPCR and immunoassays for selected factors, thereby indicating physiological effects caused by the time point of sampling. Conclusion: Prediction of potential unintended effects of GM-food/feed by transcriptome based profiling of intestinal tissue presents a novel approach to complement classical toxicological testing procedures. Including the detection of alterations in signaling pathways in toxicity testing procedures may enhance the confidence in outcomes of toxicological trials. In this study, no significant GM-related changes in intestinal expression profiles were found in rats fed GM-maize MON810. Relevant alterations of selected cellular pathways (apoptosis, DNA damage and repair, UPR) pointing toward intestinal toxicity of the diets were not observed. Transcriptomic profiles did not reveal perturbations of pathways associated with toxicity, underlining the study results revealed by classical OECD endpoints.
Collapse
Affiliation(s)
- Jutta Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Marc Bohmer
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Nils Bohmer
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Christina Backes
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Pablo Steinberg
- Institute for Food Toxicology and Analytical Chemistry, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Dagmar Zeljenková
- Faculty of Public Health, Slovak Medical University in Bratislava, Bratislava, Slovakia
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
25
|
Yi-lin L, Ke Z, Dan W, Xi-hong Z, Zheng R, Xin W, Yu-long Y. Dynamic feeding low and high methionine diets affect the diurnal rhythm of amino acid transporters and clock related genes in jejunum of laying hens. BIOL RHYTHM RES 2017. [DOI: 10.1080/09291016.2017.1395531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Liu Yi-lin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zhang Ke
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Wan Dan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Zhou Xi-hong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
| | - Ruan Zheng
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Wu Xin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yin Yu-long
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha, China
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha, China
- State Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Xu L, Wu T, Li H, Ni Y, Fu Z. An individual 12-h shift of the light-dark cycle alters the pancreatic and duodenal circadian rhythm and digestive function. Acta Biochim Biophys Sin (Shanghai) 2017; 49:954-961. [PMID: 28981604 DOI: 10.1093/abbs/gmx084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Indexed: 12/28/2022] Open
Abstract
In mammals, behavioral and physiological rhythms are controlled by circadian clocks which are entrained by environmental light and food signals. However, how the environmental cues affect digestive tract's circadian clock remains poorly understood. Therefore, in order to elucidate the effect of light cue on the resetting of the peripheral clocks, we investigated the expressions of clock genes (Bmal1, Cry1, Rev-erbα, Per1, and Per2) and digestive function genes (Cck, Cck-1r, Sct, Sctr, and Ctrb1) in the pancreas and duodenum of rats after the light-dark (LD) cycle reversal for 7 days. We found that both the clock genes and digestive function genes exhibited a clear and similar daily rhythmicity in the pancreas and duodenum of rats. After reversal of the LD cycle for 7 days, the expressions of clock genes in pancreas, including Bmal1, Cry1, and Rev-erbα were affected; whereas the expression of Per1 gene failed to fit the cosine wave. However, in the duodenum the shifted genes were Bmal1, Rev-erbα, and Per2; in parallel, the Per1 gene expression also lost its circadian rhythm by reversal of the LD cycle. Therefore, the acrophases of the clock genes were shifted in a tissue- and gene-specific manner. Furthermore, the profiles of the digestive function genes, including Sctr and Ctrb1, were also affected by changes in LD cycle. These observations suggest that the mechanisms underlying the pancreatic and duodenal clocks are distinct, and there may be a potential linkage between the circadian clock system and the digestive system.
Collapse
Affiliation(s)
- Liang Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Tao Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Haifeng Li
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
27
|
McAlpine CS, Swirski FK. Circadian Influence on Metabolism and Inflammation in Atherosclerosis. Circ Res 2017; 119:131-41. [PMID: 27340272 DOI: 10.1161/circresaha.116.308034] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/11/2016] [Indexed: 11/16/2022]
Abstract
Many aspects of human health and disease display daily rhythmicity. The brain's suprachiasmic nucleus, which interprets recurring external stimuli, and autonomous molecular networks in peripheral cells together, set our biological circadian clock. Disrupted or misaligned circadian rhythms promote multiple pathologies including chronic inflammatory and metabolic diseases such as atherosclerosis. Here, we discuss studies suggesting that circadian fluctuations in the vessel wall and in the circulation contribute to atherogenesis. Data from humans and mice indicate that an impaired molecular clock, disturbed sleep, and shifting light-dark patterns influence leukocyte and lipid supply in the circulation and alter cellular behavior in atherosclerotic lesions. We propose that a better understanding of both local and systemic circadian rhythms in atherosclerosis will enhance clinical management, treatment, and public health policy.
Collapse
Affiliation(s)
- Cameron S McAlpine
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston.
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
28
|
Mu C, Yang Y, Zhu W. Gut Microbiota: The Brain Peacekeeper. Front Microbiol 2016; 7:345. [PMID: 27014255 PMCID: PMC4794499 DOI: 10.3389/fmicb.2016.00345] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 03/04/2016] [Indexed: 12/15/2022] Open
Abstract
Gut microbiota regulates intestinal and extraintestinal homeostasis. Accumulating evidence suggests that the gut microbiota may also regulate brain function and behavior. Results from animal models indicate that disturbances in the composition and functionality of some microbiota members are associated with neurophysiological disorders, strengthening the idea of a microbiota–gut–brain axis and the role of microbiota as a “peacekeeper” in the brain health. Here, we review recent discoveries on the role of the gut microbiota in central nervous system-related diseases. We also discuss the emerging concept of the bidirectional regulation by the circadian rhythm and gut microbiota, and the potential role of the epigenetic regulation in neuronal cell function. Microbiome studies are also highlighted as crucial in the development of targeted therapies for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Chunlong Mu
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University Nanjing, China
| | - Yuxiang Yang
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University Nanjing, China
| | - Weiyun Zhu
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology, Nanjing Agricultural University Nanjing, China
| |
Collapse
|
29
|
Abstract
Robust circadian rhythms in metabolic processes have been described in both humans and animal models, at the whole body, individual organ, and even cellular level. Classically, these time-of-day-dependent rhythms have been considered secondary to fluctuations in energy/nutrient supply/demand associated with feeding/fasting and wake/sleep cycles. Renewed interest in this field has been fueled by studies revealing that these rhythms are driven, at least in part, by intrinsic mechanisms and that disruption of metabolic synchrony invariably increases the risk of cardiometabolic disease. The objectives of this paper are to provide a comprehensive review regarding rhythms in glucose, lipid, and protein/amino acid metabolism, the relative influence of extrinsic (eg, neurohumoral factors) versus intrinsic (eg, cell autonomous circadian clocks) mediators, the physiologic roles of these rhythms in terms of daily fluctuations in nutrient availability and activity status, as well as the pathologic consequences of dyssynchrony.
Collapse
Affiliation(s)
- Graham R McGinnis
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
30
|
Kumar Jha P, Challet E, Kalsbeek A. Circadian rhythms in glucose and lipid metabolism in nocturnal and diurnal mammals. Mol Cell Endocrinol 2015; 418 Pt 1:74-88. [PMID: 25662277 DOI: 10.1016/j.mce.2015.01.024] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/12/2015] [Accepted: 01/19/2015] [Indexed: 12/22/2022]
Abstract
Most aspects of energy metabolism display clear variations during day and night. This daily rhythmicity of metabolic functions, including hormone release, is governed by a circadian system that consists of the master clock in the suprachiasmatic nuclei of the hypothalamus (SCN) and many secondary clocks in the brain and peripheral organs. The SCN control peripheral timing via the autonomic and neuroendocrine system, as well as via behavioral outputs. The sleep-wake cycle, the feeding/fasting rhythm and most hormonal rhythms, including that of leptin, ghrelin and glucocorticoids, usually show an opposite phase (relative to the light-dark cycle) in diurnal and nocturnal species. By contrast, the SCN clock is most active at the same astronomical times in these two categories of mammals. Moreover, in both species, pineal melatonin is secreted only at night. In this review we describe the current knowledge on the regulation of glucose and lipid metabolism by central and peripheral clock mechanisms. Most experimental knowledge comes from studies in nocturnal laboratory rodents. Nevertheless, we will also mention some relevant findings in diurnal mammals, including humans. It will become clear that as a consequence of the tight connections between the circadian clock system and energy metabolism, circadian clock impairments (e.g., mutations or knock-out of clock genes) and circadian clock misalignments (such as during shift work and chronic jet-lag) have an adverse effect on energy metabolism, that may trigger or enhancing obese and diabetic symptoms.
Collapse
Affiliation(s)
- Pawan Kumar Jha
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France; International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| | - Etienne Challet
- Regulation of Circadian Clocks Team, Institute of Cellular and Integrative Neurosciences, UPR3212, Centre National de la Recherche Scientifique (CNRS), University of Strasbourg, France; International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; International Associated Laboratory LIA1061 Understanding the Neural Basis of Diurnality, CNRS, France and the Netherlands; Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Dridi I, Ben-Cherif W, Haouas Z, Aouam K, Ben-Attia M, Reinberg A, Boughattas NA. Gastrointestinal toxicity of mycophenolate mofetil in rats: Effect of administration time. Chronobiol Int 2015; 32:1373-84. [DOI: 10.3109/07420528.2015.1082481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Abstract
Various intestinal functions exhibit circadian rhythmicity. Disruptions in these rhythms as in shift workers and transcontinental travelers are associated with intestinal discomfort. Circadian rhythms are controlled at the molecular level by core clock and clock-controlled genes. These clock genes are expressed in intestinal cells, suggesting that they might participate in the circadian regulation of intestinal functions. A major function of the intestine is nutrient absorption. Here, we will review absorption of proteins, carbohydrates, and lipids and circadian regulation of various transporters involved in their absorption. A better understanding of circadian regulation of intestinal absorption might help control several metabolic disorders and attenuate intestinal discomfort associated with disruptions in sleep-wake cycles.
Collapse
Affiliation(s)
- M Mahmood Hussain
- Department of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, New York, USA, and VA New York Harbor Healthcare System, Brooklyn, New York, USA
| | - Xiaoyue Pan
- Department of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, New York, USA, and VA New York Harbor Healthcare System, Brooklyn, New York, USA
| |
Collapse
|
33
|
Abstract
Plasma levels of triacylglycerols and diacylglycerols, the lipoproteins that transport them, and proteins involved in their absorption from the intestinal lumen fluctuate in a circadian manner. These changes are likely controlled by clock genes expressed in the intestine that are probably synchronized by neuronal and humoral signals from the suprachiasmatic nuclei, which constitute a master clock entrained by light signals from the eyes and from the environment, e.g., food availability. Acute changes in circadian rhythms--e.g., due to nonsynchronous work schedules or a transcontinental flight--may trigger intestinal discomfort. Chronic disruptions in circadian control mechanisms may predispose the individual to irritable bowel syndrome, gastroesophageal reflux disease, and peptic ulcer disease. A more detailed understanding of the molecular mechanisms underlying temporal changes in intestinal activity might allow us to identify novel targets for developing therapeutic approaches to these disorders.
Collapse
Affiliation(s)
- M Mahmood Hussain
- Departments of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, New York 11203, and Virginia New York Harbor Healthcare System, Brooklyn, New York 11209;
| |
Collapse
|
34
|
The circadian clock maintains cardiac function by regulating mitochondrial metabolism in mice. PLoS One 2014; 9:e112811. [PMID: 25389966 PMCID: PMC4229239 DOI: 10.1371/journal.pone.0112811] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 10/15/2014] [Indexed: 01/20/2023] Open
Abstract
Cardiac function is highly dependent on oxidative energy, which is produced by mitochondrial respiration. Defects in mitochondrial function are associated with both structural and functional abnormalities in the heart. Here, we show that heart-specific ablation of the circadian clock gene Bmal1 results in cardiac mitochondrial defects that include morphological changes and functional abnormalities, such as reduced enzymatic activities within the respiratory complex. Mice without cardiac Bmal1 function show a significant decrease in the expression of genes associated with the fatty acid oxidative pathway, the tricarboxylic acid cycle, and the mitochondrial respiratory chain in the heart and develop severe progressive heart failure with age. Importantly, similar changes in gene expression related to mitochondrial oxidative metabolism are also observed in C57BL/6J mice subjected to chronic reversal of the light-dark cycle; thus, they show disrupted circadian rhythmicity. These findings indicate that the circadian clock system plays an important role in regulating mitochondrial metabolism and thereby maintains cardiac function.
Collapse
|
35
|
Oosterman JE, Foppen E, van der Spek R, Fliers E, Kalsbeek A, la Fleur SE. Timing of fat and liquid sugar intake alters substrate oxidation and food efficiency in male Wistar rats. Chronobiol Int 2014; 32:289-98. [PMID: 25317718 DOI: 10.3109/07420528.2014.971177] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In addition to the amount of ingested calories, both timing of food intake and meal composition are determinants of body weight gain. However, at present, it is unknown if the inappropriate timing of diet components is responsible for body weight gain. In the present study, we therefore studied a time-dependent effect of the diet composition on energy homeostasis. Male Wistar rats were subjected to chow ad libitum (chow group) or a choice diet with saturated fat, a 30% sugar solution, chow and tap water. The choice diet was provided either with all components ad libitum (AL), with ad libitum access to chow, tap water and a 30% sugar solution, but with access to saturated fat only during the light period (LF), or with ad libitum access to chow, tap water and saturated fat, but access to a 30% sugar solution only during the light period (LS). Caloric intake and body weight gain were monitored during 31 days. Energy expenditure was measured in the third week in calorimetric cages. All rats on a choice diet showed hyperphagia and gained more body weight compared to the chow group. Within the choice diet groups, rats on the LS diet were most food efficient (i.e. gained most body weight per ingested calorie) and showed a lower respiratory exchange ratio (RER) with an anti-phasic pattern, whereas no differences in locomotor activity or heat production were found. Collectively these data indicate that the timing of the diet composition affects food efficiency, most likely due to a shifted oxidation pattern, which can predispose for obesity. Further studies are underway to assess putative mechanisms involved in this dysregulation.
Collapse
Affiliation(s)
- Johanneke E Oosterman
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands and
| | | | | | | | | | | |
Collapse
|
36
|
Rothschild J, Lagakos W. Implications of enteral and parenteral feeding times: considering a circadian picture. JPEN J Parenter Enteral Nutr 2014; 39:266-70. [PMID: 25239111 DOI: 10.1177/0148607114551026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
37
|
Abstract
Among all the metabolites present in the plasma, lipids, mainly triacylglycerol and diacylglycerol, show extensive circadian rhythms. These lipids are transported in the plasma as part of lipoproteins. Lipoproteins are synthesized primarily in the liver and intestine and their production exhibits circadian rhythmicity. Studies have shown that various proteins involved in lipid absorption and lipoprotein biosynthesis show circadian expression. Further, intestinal epithelial cells express circadian clock genes and these genes might control circadian expression of different proteins involved in intestinal lipid absorption. Intestinal circadian clock genes are synchronized by signals emanating from the suprachiasmatic nuclei that constitute a master clock and from signals coming from other environmental factors, such as food availability. Disruptions in central clock, as happens due to disruptions in the sleep/wake cycle, affect intestinal function. Similarly, irregularities in temporal food intake affect intestinal function. These changes predispose individuals to various metabolic disorders, such as metabolic syndrome, obesity, diabetes, and atherosclerosis. Here, we summarize how circadian rhythms regulate microsomal triglyceride transfer protein, apoAIV, and nocturnin to affect diurnal regulation of lipid absorption.
Collapse
Affiliation(s)
- M Mahmood Hussain
- Departments of Cell Biology and Pediatrics, State University of New York Downstate Medical Center, Brooklyn, NY 11203; and Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209
| | - Xiaoyue Pan
- Departments of Cell Biology and Pediatrics, State University of New York Downstate Medical Center, Brooklyn, NY 11203; and Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209
| |
Collapse
|
38
|
Moore SR, Pruszka J, Vallance J, Aihara E, Matsuura T, Montrose MH, Shroyer NF, Hong CI. Robust circadian rhythms in organoid cultures from PERIOD2::LUCIFERASE mouse small intestine. Dis Model Mech 2014; 7:1123-30. [PMID: 24997189 PMCID: PMC4142732 DOI: 10.1242/dmm.014399] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Disruption of circadian rhythms is a risk factor for several human gastrointestinal (GI) diseases, ranging from diarrhea to ulcers to cancer. Four-dimensional tissue culture models that faithfully mimic the circadian clock of the GI epithelium would provide an invaluable tool to understand circadian regulation of GI health and disease. We hypothesized that rhythmicity of a key circadian component, PERIOD2 (PER2), would diminish along a continuum from ex vivo intestinal organoids (epithelial ‘miniguts’), nontransformed mouse small intestinal epithelial (MSIE) cells and transformed human colorectal adenocarcinoma (Caco-2) cells. Here, we show that bioluminescent jejunal explants from PERIOD2::LUCIFERASE (PER2::LUC) mice displayed robust circadian rhythms for >72 hours post-excision. Circadian rhythms in primary or passaged PER2::LUC jejunal organoids were similarly robust; they also synchronized upon serum shock and persisted beyond 2 weeks in culture. Remarkably, unshocked organoids autonomously synchronized rhythms within 12 hours of recording. The onset of this autonomous synchronization was slowed by >2 hours in the presence of the glucocorticoid receptor antagonist RU486 (20 μM). Doubling standard concentrations of the organoid growth factors EGF, Noggin and R-spondin enhanced PER2 oscillations, whereas subtraction of these factors individually at 24 hours following serum shock produced no detectable effects on PER2 oscillations. Growth factor pulses induced modest phase delays in unshocked, but not serum-shocked, organoids. Circadian oscillations of PER2::LUC bioluminescence aligned with Per2 mRNA expression upon analysis using quantitative PCR. Concordant findings of robust circadian rhythms in bioluminescent jejunal explants and organoids provide further evidence for a peripheral clock that is intrinsic to the intestinal epithelium. The rhythmic and organotypic features of organoids should offer unprecedented advantages as a resource for elucidating the role of circadian rhythms in GI stem cell dynamics, epithelial homeostasis and disease.
Collapse
Affiliation(s)
- Sean R Moore
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH 45229-3039, USA
| | - Jill Pruszka
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH 45229-3039, USA
| | - Jefferson Vallance
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH 45229-3039, USA
| | - Eitaro Aihara
- Molecular and Cellular Physiology, University of Cincinnati College of Medicine, OH 45267-0576, USA
| | - Toru Matsuura
- Molecular and Cellular Physiology, University of Cincinnati College of Medicine, OH 45267-0576, USA
| | - Marshall H Montrose
- Molecular and Cellular Physiology, University of Cincinnati College of Medicine, OH 45267-0576, USA
| | - Noah F Shroyer
- Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, University of Cincinnati, OH 45229-3039, USA
| | - Christian I Hong
- Molecular and Cellular Physiology, University of Cincinnati College of Medicine, OH 45267-0576, USA
| |
Collapse
|
39
|
Adamovich Y, Rousso-Noori L, Zwighaft Z, Neufeld-Cohen A, Golik M, Kraut-Cohen J, Wang M, Han X, Asher G. Circadian clocks and feeding time regulate the oscillations and levels of hepatic triglycerides. Cell Metab 2014; 19:319-30. [PMID: 24506873 PMCID: PMC4261230 DOI: 10.1016/j.cmet.2013.12.016] [Citation(s) in RCA: 274] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/12/2013] [Accepted: 12/16/2013] [Indexed: 11/26/2022]
Abstract
Circadian clocks play a major role in orchestrating daily physiology, and their disruption can evoke metabolic diseases such as fatty liver and obesity. To study the role of circadian clocks in lipid homeostasis, we performed an extensive lipidomic analysis of liver tissues from wild-type and clock-disrupted mice either fed ad libitum or night fed. To our surprise, a similar fraction of lipids (∼17%) oscillated in both mouse strains, most notably triglycerides, but with completely different phases. Moreover, several master lipid regulators (e.g., PPARα) and enzymes involved in triglyceride metabolism retained their circadian expression in clock-disrupted mice. Nighttime restricted feeding shifted the phase of triglyceride accumulation and resulted in ∼50% decrease in hepatic triglyceride levels in wild-type mice. Our findings suggest that circadian clocks and feeding time dictate the phase and levels of hepatic triglyceride accumulation; however, oscillations in triglycerides can persist in the absence of a functional clock.
Collapse
Affiliation(s)
- Yaarit Adamovich
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liat Rousso-Noori
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ziv Zwighaft
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adi Neufeld-Cohen
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Marina Golik
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Judith Kraut-Cohen
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Miao Wang
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA
| | - Xianlin Han
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA
| | - Gad Asher
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
40
|
Nakamura Y, Andrés F, Kanehara K, Liu YC, Coupland G, Dörmann P. Diurnal and circadian expression profiles of glycerolipid biosynthetic genes in Arabidopsis. PLANT SIGNALING & BEHAVIOR 2014; 9:e29715. [PMID: 25763705 PMCID: PMC4205134 DOI: 10.4161/psb.29715] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Glycerolipid composition in plant membranes oscillates in response to diurnal change. However, its functional significance remained unclear. A recent discovery that Arabidopsis florigen FT binds diurnally oscillating phosphatidylcholine molecules to promote flowering suggests that diurnal oscillation of glycerolipid composition is an important input in flowering time control. Taking advantage of public microarray data, we globally analyzed the expression pattern of glycerolipid biosynthetic genes in Arabidopsis under long-day, short-day, and continuous light conditions. The results revealed that 12 genes associated with glycerolipid metabolism showed significant oscillatory profiles. Interestingly, expression of most of these genes followed circadian profiles, suggesting that glycerolipid biosynthesis is partially under clock regulation. The oscillating expression profile of one representative gene, PECT1, was analyzed in detail. Expression of PECT1 showed a circadian pattern highly correlated with that of the clock-regulated gene GIGANTEA. Thus, our study suggests that a considerable number of glycerolipid biosynthetic genes are under circadian control.
Collapse
Affiliation(s)
- Yuki Nakamura
- Institute of Plant and Microbial Biology; Academia Sinica; Nankang, Taipei, Taiwan
- Correspondence to: Yuki Nakamura,
| | - Fernando Andrés
- Max-Planck-Institute for Plant Breeding Research; Cologne, Germany
| | - Kazue Kanehara
- Institute of Plant and Microbial Biology; Academia Sinica; Nankang, Taipei, Taiwan
| | - Yu-chi Liu
- Institute of Plant and Microbial Biology; Academia Sinica; Nankang, Taipei, Taiwan
| | - George Coupland
- Max-Planck-Institute for Plant Breeding Research; Cologne, Germany
| | - Peter Dörmann
- Institute of Molecular Physiology and Biotechnology of Plants; University of Bonn; Bonn, Germany
| |
Collapse
|
41
|
Abstract
BACKGROUND Clock is a key transcription factor that positively controls circadian regulation. However, its role in plasma cholesterol homeostasis and atherosclerosis has not been studied. METHODS AND RESULTS We show for the first time that dominant-negative Clock mutant protein (Clock(Δ19/Δ19)) enhances plasma cholesterol and atherosclerosis in 3 different mouse models. Detailed analyses revealed that Clk(Δ19/Δ19)Apoe(-/-) mice display hypercholesterolemia resulting from the accumulation of apolipoprotein B48-containing cholesteryl ester-rich lipoproteins. Physiological studies showed that enhanced cholesterol absorption by the intestine contributes to hypercholesterolemia. Molecular studies indicated that the expression of Niemann Pick C1 Like 1, Acyl-CoA:Cholesterol acyltransferase 1, and microsomal triglyceride transfer protein in the intestines of Clk(Δ19/Δ19)Apoe(-/-) mice was high and that enterocytes assembled and secreted more chylomicrons. Furthermore, we identified macrophage dysfunction as another potential cause of increased atherosclerosis in Clk(Δ19/Δ19)Apoe(-/-) mice. Macrophages from Clk(Δ19/Δ19)Apoe(-/-) mice expressed higher levels of scavenger receptors and took up more modified lipoproteins compared with Apoe(-/-) mice, but they expressed low levels of ATP binding casette protein family A member 1 and were defective in cholesterol efflux. Molecular studies revealed that Clock regulates ATP binding casette protein family A member 1 expression in macrophages by modulating upstream transcription factor 2 expression. CONCLUSIONS Clock(Δ19/Δ19) protein enhances atherosclerosis by increasing intestinal cholesterol absorption, augmenting uptake of modified lipoproteins by macrophages, and reducing cholesterol efflux from macrophages. These studies establish that circadian Clock activity is crucial in maintaining low plasma cholesterol levels and in reducing atherogenesis in mice.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Departments of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, NY
| | | | | |
Collapse
|
42
|
Abstract
A new study has identified the sophisticated communication between the gut microbiota and intestinal epithelial cells that controls diurnal variations in the secretion of corticosteroids in mice. The absence of gut bacteria disrupts this communication and contributes to hypertriglyceridaemia, hyperglycaemia and insulin resistance.
Collapse
|
43
|
Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell 2013; 153:812-27. [PMID: 23663780 DOI: 10.1016/j.cell.2013.04.020] [Citation(s) in RCA: 414] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/24/2012] [Accepted: 04/04/2013] [Indexed: 12/15/2022]
Abstract
Alterations of symbiosis between microbiota and intestinal epithelial cells (IEC) are associated with intestinal and systemic pathologies. Interactions between bacterial products (MAMPs) and Toll-like receptors (TLRs) are known to be mandatory for IEC homeostasis, but how TLRs may time homeostatic functions with circadian changes is unknown. Our functional and molecular dissections of the IEC circadian clock demonstrate that its integrity is required for microbiota-IEC dialog. In IEC, the antiphasic expression of the RORα activator and RevErbα repressor clock output regulators generates a circadian rhythmic TLR expression that converts the temporally arrhythmic microbiota signaling into circadian rhythmic JNK and IKKβ activities, which prevents RevErbα activation by PPARα that would disrupt the circadian clock. Moreover, through activation of AP1 and NF-κB, these activities, together with RORα and RevErbα, enable timing homeostatic functions of numerous genes with IEC circadian events. Interestingly, microbiota signaling deficiencies induce a prediabetic syndrome due to ileal corticosterone overproduction consequent to clock disruption.
Collapse
|
44
|
Soh J, Iqbal J, Queiroz J, Fernandez-Hernando C, Hussain MM. MicroRNA-30c reduces hyperlipidemia and atherosclerosis in mice by decreasing lipid synthesis and lipoprotein secretion. Nat Med 2013; 19:892-900. [PMID: 23749231 PMCID: PMC4121125 DOI: 10.1038/nm.3200] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 04/16/2013] [Indexed: 02/07/2023]
Abstract
Hyperlipidemia is a risk factor for various cardiovascular and metabolic disorders. Overproduction of lipoproteins, a process critically dependent on microsomal triglyceride transfer protein (MTP), can contribute to hyperlipidemia. We show that microRNA-30c (miR-30c) interacts with the 3′-untranslated region of the MTP mRNA and induces degradation leading to reductions in its activity and media apolipoprotein B. Further, miR-30c reduces hyperlipidemia and atherosclerosis in Western diet fed mice by decreasing lipid synthesis and secretion of triglyceride-rich apoB-containing lipoproteins. Therefore, miR-30c coordinately reduces lipid biosynthesis and lipoprotein secretion to control hepatic and plasma lipids and might be useful in treating hyperlipidemias and associated disorders.
Collapse
Affiliation(s)
- James Soh
- School of Graduate Studies, Molecular and Cell Biology Program, State University of New York Downstate Medical Center, Brooklyn, New York, USA
| | | | | | | | | |
Collapse
|
45
|
Pan X, Munshi MK, Iqbal J, Queiroz J, Sirwi AA, Shah S, Younus A, Hussain MM. Circadian regulation of intestinal lipid absorption by apolipoprotein AIV involves forkhead transcription factors A2 and O1 and microsomal triglyceride transfer protein. J Biol Chem 2013; 288:20464-76. [PMID: 23729668 DOI: 10.1074/jbc.m113.473454] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have shown previously that Clock, microsomal triglyceride transfer protein (MTP), and nocturnin are involved in the circadian regulation of intestinal lipid absorption. Here, we clarified the role of apolipoprotein AIV (apoAIV) in the diurnal regulation of plasma lipids and intestinal lipid absorption in mice. Plasma triglyceride in apoAIV(-/-) mice showed diurnal variations similar to apoAIV(+/+) mice; however, the increases in plasma triglyceride at night were significantly lower in these mice. ApoAIV(-/-) mice absorbed fewer lipids at night and showed blunted response to daytime feeding. To explain reasons for these lower responses, we measured MTP expression; intestinal MTP was low at night, and its induction after food entrainment was less in apoAIV(-/-) mice. Conversely, apoAIV overexpression increased MTP mRNA in hepatoma cells, indicating transcriptional regulation. Mechanistic studies revealed that sequences between -204/-775 bp in the MTP promoter respond to apoAIV and that apoAIV enhances expression of FoxA2 and FoxO1 transcription factors and their binding to the identified cis elements in the MTP promoter at night. Knockdown of FoxA2 and FoxO1 abolished apoAIV-mediated MTP induction. Similarly, knockdown of apoAIV in differentiated Caco-2 cells reduced MTP, FoxA2, and FoxO1 mRNA levels, cellular MTP activity, and media apoB. Moreover, FoxA2 and FoxO1 expression showed diurnal variations, and their expression was significantly lower in apoAIV(-/-) mice. These data indicate that apoAIV modulates diurnal changes in lipid absorption by regulating forkhead transcription factors and MTP and that inhibition of apoAIV expression might reduce plasma lipids.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Gamble KL, Young ME. Metabolism as an integral cog in the mammalian circadian clockwork. Crit Rev Biochem Mol Biol 2013; 48:317-31. [PMID: 23594144 DOI: 10.3109/10409238.2013.786672] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Circadian rhythms are an integral part of life. These rhythms are apparent in virtually all biological processes studies to date, ranging from the individual cell (e.g. DNA synthesis) to the whole organism (e.g. behaviors such as physical activity). Oscillations in metabolism have been characterized extensively in various organisms, including mammals. These metabolic rhythms often parallel behaviors such as sleep/wake and fasting/feeding cycles that occur on a daily basis. What has become increasingly clear over the past several decades is that many metabolic oscillations are driven by cell-autonomous circadian clocks, which orchestrate metabolic processes in a temporally appropriate manner. During the process of identifying the mechanisms by which clocks influence metabolism, molecular-based studies have revealed that metabolism should be considered an integral circadian clock component. The implications of such an interrelationship include the establishment of a vicious cycle during cardiometabolic disease states, wherein metabolism-induced perturbations in the circadian clock exacerbate metabolic dysfunction. The purpose of this review is therefore to highlight recent insights gained regarding links between cell-autonomous circadian clocks and metabolism and the implications of clock dysfunction in the pathogenesis of cardiometabolic diseases.
Collapse
Affiliation(s)
- Karen L Gamble
- Division of Behavioral Neurobiology, Department of Psychiatry, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
47
|
Bray MS, Ratcliffe WF, Grenett MH, Brewer RA, Gamble KL, Young ME. Quantitative analysis of light-phase restricted feeding reveals metabolic dyssynchrony in mice. Int J Obes (Lond) 2012; 37:843-52. [PMID: 22907695 PMCID: PMC3505273 DOI: 10.1038/ijo.2012.137] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background Considerable evidence suggests that the time of day at which calories are consumed markedly impacts body weight gain and adiposity. However, a precise quantification of energy balance parameters during controlled animal studies enforcing time-of-day-restricted feeding is currently lacking in the absence of direct human interaction. Objective The purpose of the present study was therefore to quantify the effects of restricted feeding during the light (sleep) phase in a fully-automated, computer-controlled comprehensive laboratory animal monitoring system (CLAMS) designed to modulate food access in a time-of-day-dependent manner. Energy balance, gene expression (within metabolically-relevant tissues), humoral factors, and body weight were assessed. Results We report that relative to mice fed only during the dark (active) phase, light (sleep) phase fed mice: 1) consume a large meal upon initiation of food availability; 2) consume greater total calories per day; 3) exhibit a higher RER (indicative of decreased reliance on lipid/fatty acid oxidation); 4) exhibit tissue-specific alterations in the phases and amplitudes of circadian clock and metabolic genes in metabolically active tissues (greatest phase differences observed in the liver, and diminution of amplitudes in epididymal fat, gastrocnemius muscle, and heart); 5) exhibit diminished amplitude in humoral factor diurnal variations (e.g., corticosterone); and 6) exhibit greater weight gain within 9 days of restricted feeding. Conclusions Collectively, these data suggest that weight gain following light (sleep) phase restricted feeding is associated with significant alterations in energy balance, as well as dyssynchrony between metabolically active organs.
Collapse
Affiliation(s)
- M S Bray
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
48
|
Hussain MM, Jin W, Jiang XC. Mechanisms involved in cellular ceramide homeostasis. Nutr Metab (Lond) 2012; 9:71. [PMID: 22849442 PMCID: PMC3463440 DOI: 10.1186/1743-7075-9-71] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 07/13/2012] [Indexed: 11/10/2022] Open
Abstract
Sphingolipids are ubiquitous and critical components of biological membranes. Their biosynthesis starts with soluble precursors in the endoplasmic reticulum and culminates in the Golgi complex and plasma membrane. Ceramides are important intermediates in the biosynthesis of sphingolipids, such as sphingomyelin, and their overload in the membranes is injurious to cells. The major product of ceramide metabolism is sphingomyelin. We observed that sphingomyelin synthase (SMS) 1 or SMS2 deficiencies significantly decreased plasma and liver sphingomyelin levels. However, SMS2 but not SMS1 deficiency increased plasma ceramides. Surprisingly, SMS1 deficiency significantly increased glucosylceramide and ganglioside GM3, but SMS2 deficiency did not. To explain these unexpected findings about modest to no significant changes in ceramides and increases in other sphingolipids after the ablation of SMS1, we hypothesize that cells have evolved several organelle specific mechanisms to maintain ceramide homeostasis. First, ceramides in the endoplasmic reticulum membranes are controlled by its export to Golgi by protein mediated transfer. Second, in the Golgi, ceramide levels are modulated by their enzymatic conversion to different sphingolipids such as sphingomyelin, and glucosylceramides. Additionally, these sphingolipids can become part of triglyceride-rich apolipoprotein B-containing lipoproteins and be secreted. Third, in the plasma membrane ceramide levels are maintained by ceramide/sphingomyelin cycle, delivery to lysosomes, and efflux to extracellular plasma acceptors. All these pathways might have evolved to ensure steady cellular ceramide levels.
Collapse
Affiliation(s)
- M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Weijun Jin
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW To summarize the new knowledge about the regulation of dietary lipid absorption by circadian locomotor output cycles kaput (Clock) and Nocturnin. RECENT FINDINGS Recent findings have shown that Clock and Nocturnin, proteins involved in circadian regulation, play an important role in the regulation of dietary lipid absorption. Clock deficiency increases, whereas Nocturnin deficiency decreases lipid absorption. Clock plays a role in turning off the genes involved in lipid absorption at the onset of the day. Molecular studies revealed that Clock binds to the promoter of small heterodimer partner to enhance its transcription. When levels are high, small heterodimer partner interacts with the transcription factors associated with the promoter of microsomal triglyceride transfer protein to repress transcription. Reduced microsomal triglyceride transfer protein levels are correlated with low intestinal lipid absorption and plasma lipid levels. In contrast, Nocturnin assists in lipid absorption by regulating their partitioning in different intracellular compartments. SUMMARY Clock and Nocturnin regulate lipid absorption involving different mechanisms. It is likely that other clock genes also modulate lipid absorption and plasma lipid levels.
Collapse
Affiliation(s)
- M Mahmood Hussain
- Departments of Cell Biology and Pediatrics, SUNY Downstate Medical Center, Brooklyn, New York, USA.
| | | |
Collapse
|
50
|
Stubblefield JJ, Terrien J, Green CB. Nocturnin: at the crossroads of clocks and metabolism. Trends Endocrinol Metab 2012; 23:326-33. [PMID: 22608110 PMCID: PMC3389576 DOI: 10.1016/j.tem.2012.03.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/28/2012] [Accepted: 03/30/2012] [Indexed: 02/06/2023]
Abstract
Many aspects of metabolism exhibit daily rhythmicity under the control of endogenous circadian clocks, and disruptions in circadian timing result in dysfunctions associated with the metabolic syndrome. Nocturnin (Noc) is a robustly rhythmic gene that encodes a deadenylase thought to be involved in the removal of polyA tails from mRNAs. Mice lacking the Noc gene display resistance to diet-induced obesity and hepatic steatosis, due in part to reduced lipid trafficking in the small intestine. In addition, Noc appears to play important roles in other tissues and has been implicated in lipid metabolism, adipogenesis, glucose homeostasis, inflammation and osteogenesis. Therefore, Noc is a potential key post-transcriptional mediator in the circadian control of many metabolic processes.
Collapse
Affiliation(s)
- Jeremy J Stubblefield
- Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, NB4.204G, Dallas, TX 75390-9111, USA
| | | | | |
Collapse
|