1
|
Ma T, Matsuo R, Kurogi K, Miyamoto S, Morita T, Shinozuka M, Taniguchi F, Ikegami K, Yasuo S. Sex-dependent effects of chronic jet lag on circadian rhythm and metabolism in mice. Biol Sex Differ 2024; 15:102. [PMID: 39639385 PMCID: PMC11619446 DOI: 10.1186/s13293-024-00679-z] [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: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The circadian clock integrates external environmental changes into the internal physiology of organisms. Perturbed circadian clocks due to misaligned light cycles increase the risk of diseases, including metabolic disorders. However, the effects of sex differences in this context remain unclear. METHODS Circadian misalignment was induced by a chronic jet lag (CJL) shift schedule (light-on time advanced by 6 h every 2 days) in C57BL/6N male and female mice. Core body temperature and activity rhythms were recorded using a nano tag, and the gene expression rhythms of clock and clock-controlled genes in the liver and adrenal glands were analyzed using qPCR. Glucose metabolism and insulin response were evaluated using glucose tolerance, insulin sensitivity, and glucose response assays. Castration and testosterone replacement were performed to assess the fundamental role of testosterone in male phenotypes under CJL. RESULTS Under CJL treatment, male mice exhibited increased weight gain, whereas females exhibited decreased weight gain compared to that of the respective controls. CJL treatment induced a lower robustness of circadian rhythms in core body temperature and a weaker rhythm of clock gene expression in the liver and adrenal glands in females, but not in males. Only male mice exhibited glucose intolerance under CJL conditions, without the development of insulin resistance. Castrated mice without testosterone exhibited decreased weight gain and reduced robustness of body temperature rhythm, as observed in intact females. Testosterone replacement in castrated mice recovered the CJL-induced weight gain, robustness of temperature rhythm, and glucose intolerance observed in intact males. CONCLUSIONS Significant sex-based differences were observed in circadian clock organization and metabolism under CJL. Testosterone plays a crucial role in maintaining the circadian clock and regulating CJL metabolism in males.
Collapse
Affiliation(s)
- Tiantian Ma
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Ryohei Matsuo
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Kaito Kurogi
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shunsuke Miyamoto
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Tatsumi Morita
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Marina Shinozuka
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Fuka Taniguchi
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Keisuke Ikegami
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shinobu Yasuo
- Laboratory of Regulation in Metabolism and Behavior, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
2
|
Ledesma-Aparicio J, Mailloux-Salinas P, Arias-Chávez DJ, Campos-Pérez E, Calixto-Tlacomulco S, Cruz-Rangel A, Reyes-Grajeda JP, Bravo G. Transcriptomic Analysis of the Protective Effect of Piperine on Orlistat Hepatotoxicity in Obese Male Wistar Rats. J Biochem Mol Toxicol 2024; 38:e70040. [PMID: 39503200 DOI: 10.1002/jbt.70040] [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: 03/06/2024] [Revised: 05/17/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024]
Abstract
Obesity is a risk factor for the development of noncommunicable diseases that impair the quality of life. Orlistat is one of the most widely used drugs in the management of obesity due to its accessibility and low cost. However, cases of hepatotoxicity have been reported due to the consumption of this drug. On the other hand, piperine is an alkaloid found in black pepper that has demonstrated antiobesity, antihyperlipidemic, antioxidant, prebiotic, and hepatoprotective effects. The aim of this study was to evaluate the protective effect of piperine on the toxicity of orlistat in liver tissue. Obese male rats were administered piperine (30 mg/kg), orlistat (60 mg/kg), and the orlistat-piperine combination (30 mg/kg + 60 mg/kg) daily for 6 weeks. It was observed that the orlistat-piperine treatment resulted in greater weight loss, decreased biochemical markers (lipid profile, liver enzymes, pancreatic lipase activity), and histopathological analysis showed decreased hepatic steatosis and reduction of duodenal inflammation. Transcriptomic analysis revealed that the administration of piperine with orlistat increased the expression of genes related to the beta-oxidation of fatty acids, carbohydrate metabolism, detoxification of xenobiotics, and response to oxidative stress. Therefore, the results suggest that the administration of orlistat-piperine activates signaling pathways that confer a hepatoprotective effect, reducing the toxic impact of this drug.
Collapse
Affiliation(s)
- Jessica Ledesma-Aparicio
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Patrick Mailloux-Salinas
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - David Julian Arias-Chávez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Elihu Campos-Pérez
- Departamento de Patología, Hospital General Dra Matilde Petra Montoya Lafragua, ISSSTE, Mexico City, Mexico
| | - Sandra Calixto-Tlacomulco
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Armando Cruz-Rangel
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Juan Pablo Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Guadalupe Bravo
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| |
Collapse
|
3
|
Singh A, Anjum B, Naz Q, Raza S, Sinha RA, Ahmad MK, Mehdi AA, Verma N. Night shift-induced circadian disruption: links to initiation of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis and risk of hepatic cancer. HEPATOMA RESEARCH 2024:2394-5079.2024.88. [PMID: 39525867 PMCID: PMC7616786 DOI: 10.20517/2394-5079.2024.88] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The circadian system plays a crucial role in regulating metabolic homeostasis at both systemic and tissue levels by synchronizing the central and peripheral clocks with exogenous time cues, known as zeitgebers (such as the light/dark cycle). Our body's behavioral rhythms, including sleep-wake cycles and feeding-fasting patterns, align with these extrinsic time cues. The body cannot effectively rest and repair itself when circadian rhythms are frequently disrupted. In many shift workers, the internal rhythms fail to fully synchronize with the end and start times of their shifts. Additionally, exposure to artificial light at night (LAN), irregular eating patterns, and sleep deprivation contribute to circadian disruption and misalignment. Shift work and jet lag disrupt the normal circadian rhythm of liver activity, resulting in a condition known as "circadian disruption". This disturbance adversely affects the metabolism and homeostasis of the liver, contributing to excessive fat accumulation and abnormal liver function. Additionally, extended working hours, such as prolonged night shifts, may worsen the progression of non-alcoholic fatty liver disease (NAFLD) toward non-alcoholic steatohepatitis (NASH) and increase disease severity. Studies have demonstrated a positive correlation between night shift work (NSW) and elevated liver enzymes, indicative of hepatic metabolic dysfunction, potentially increasing the risk of hepatocellular carcinoma (HCC) related to NAFLD. This review consolidates research findings on circadian disruption caused by NSW, late chronotype, jet lag, and social jet lag, drawing insights from studies involving both humans and animal models that investigate the effects of these factors on circadian rhythms in liver metabolism.
Collapse
Affiliation(s)
- Anjali Singh
- Department of Physiology, King George’s Medical University, Lucknow226003, India
| | - Baby Anjum
- Department of Neurology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Qulsoom Naz
- Department of Medicine, King George’s Medical University, Lucknow226003, India
| | - Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | - Rohit A. Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow226014, India
| | | | | | - Narsingh Verma
- Hind Institute of Medical Sciences, Sitapur 261304, India
| |
Collapse
|
4
|
Rajan PK, Udoh UAS, Finley R, Pierre SV, Sanabria J. The Biological Clock of Liver Metabolism in Metabolic Dysfunction-Associated Steatohepatitis Progression to Hepatocellular Carcinoma. Biomedicines 2024; 12:1961. [PMID: 39335475 PMCID: PMC11428469 DOI: 10.3390/biomedicines12091961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Circadian rhythms are endogenous behavioral or physiological cycles that are driven by a daily biological clock that persists in the absence of geophysical or environmental temporal cues. Circadian rhythm-related genes code for clock proteins that rise and fall in rhythmic patterns driving biochemical signals of biological processes from metabolism to physiology and behavior. Clock proteins have a pivotal role in liver metabolism and homeostasis, and their disturbances are implicated in various liver disease processes. Encoded genes play critical roles in the initiation and progression of metabolic dysfunction-associated steatohepatitis (MASH) to hepatocellular carcinoma (HCC) and their proteins may become diagnostic markers as well as therapeutic targets. Understanding molecular and metabolic mechanisms underlying circadian rhythms will aid in therapeutic interventions and may have broader clinical applications. The present review provides an overview of the role of the liver's circadian rhythm in metabolic processes in health and disease, emphasizing MASH progression and the oncogenic associations that lead to HCC.
Collapse
Affiliation(s)
- Pradeep Kumar Rajan
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Utibe-Abasi S Udoh
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Robert Finley
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
| | - Juan Sanabria
- Marshall Institute for Interdisciplinary Research, Huntington, WV 25703, USA
- Department of Surgery, School of Medicine, Marshall University, Huntington, WV 25701, USA
- Department of Nutrition and Metabolomic Core Facility, School of Medicine, Case Western Reserve University, Cleveland, OH 44100, USA
| |
Collapse
|
5
|
Koh YC, Yao CH, Lee PS, Nagabhushanam K, Ho CT, Pan MH. Hepatoprotective effect of dietary pterostilbene against high-fat-diet-induced lipid accumulation exacerbated by chronic jet lag via SIRT1 and SIRT3 activation. Phytother Res 2024; 38:4099-4113. [PMID: 38899498 DOI: 10.1002/ptr.8262] [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: 01/10/2024] [Revised: 05/19/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
Hepatic lipid metabolism is modulated by the circadian rhythm; therefore, circadian disruption may promote obesity and hepatic lipid accumulation. This study aims to investigate dietary pterostilbene (PSB) 's protective effect against high-fat-diet (HFD)-induced lipid accumulation exacerbated by chronic jet lag and the potential role of gut microbiota therein. Mice were treated with a HFD and chronic jet lag for 14 weeks. The experimental group was supplemented with 0.25% (w/w) PSB in its diet to evaluate whether PSB had a beneficial effect. Our study found that chronic jet lag exacerbates HFD-induced obesity and hepatic lipid accumulation, but these adverse effects were significantly mitigated by PSB supplementation. Specifically, PSB promoted hepatic lipolysis and β-oxidation by upregulating SIRT1 expression, which indirectly reduced oxidative stress caused by lipid accumulation. Additionally, the PSB-induced elevation of SIRT1 and SIRT3 expression helped prevent excessive autophagy and mitochondrial fission by activating Nrf2-mediated antioxidant enzymes. The result was evidenced by the use of SIRT1 and SIRT3 inhibitors in in vitro studies, which demonstrated that activation of SIRT1 and SIRT3 by PSB is crucial for the translocation of PGC-1α and Nrf2, respectively. Moreover, the analysis of gut microbiota suggested that PSB's beneficial effects were partly due to its positive modulation of gut microbial composition and functionality. The findings of this study suggest the potential of dietary PSB as a candidate to improve hepatic lipid metabolism via several mechanisms. It may be developed as a treatment adjuvant in the future.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | - Ching-Hui Yao
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | - Pei-Sheng Lee
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | | | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA
| | - Min-Hsiung Pan
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung City, Taiwan
| |
Collapse
|
6
|
Pacheco-Bernal I, Becerril-Pérez F, Bustamante-Zepeda M, González-Suárez M, Olmedo-Suárez MA, Hernández-Barrientos LR, Alarcón-Del-Carmen A, Escalante-Covarrubias Q, Mendoza-Viveros L, Hernández-Lemus E, León-Del-Río A, de la Rosa-Velázquez IA, Orozco-Solis R, Aguilar-Arnal L. Transitions in chromatin conformation shaped by fatty acids and the circadian clock underlie hepatic transcriptional reorganization in obese mice. Cell Mol Life Sci 2024; 81:309. [PMID: 39060446 PMCID: PMC11335233 DOI: 10.1007/s00018-024-05364-3] [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: 02/15/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
The circadian clock system coordinates metabolic, physiological, and behavioral functions across a 24-h cycle, crucial for adapting to environmental changes. Disruptions in circadian rhythms contribute to major metabolic pathologies like obesity and Type 2 diabetes. Understanding the regulatory mechanisms governing circadian control is vital for identifying therapeutic targets. It is well characterized that chromatin remodeling and 3D structure at genome regulatory elements contributes to circadian transcriptional cycles; yet the impact of rhythmic chromatin topology in metabolic disease is largely unexplored. In this study, we explore how the spatial configuration of the genome adapts to diet, rewiring circadian transcription and contributing to dysfunctional metabolism. We describe daily fluctuations in chromatin contacts between distal regulatory elements of metabolic control genes in livers from lean and obese mice and identify specific lipid-responsive regions recruiting the clock molecular machinery. Interestingly, under high-fat feeding, a distinct interactome for the clock-controlled gene Dbp strategically promotes the expression of distal metabolic genes including Fgf21. Alongside, new chromatin loops between regulatory elements from genes involved in lipid metabolism control contribute to their transcriptional activation. These enhancers are responsive to lipids through CEBPβ, counteracting the circadian repressor REVERBa. Our findings highlight the intricate coupling of circadian gene expression to a dynamic nuclear environment under high-fat feeding, supporting a temporally regulated program of gene expression and transcriptional adaptation to diet.
Collapse
Affiliation(s)
- Ignacio Pacheco-Bernal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Fernando Becerril-Pérez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Marcia Bustamante-Zepeda
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Mirna González-Suárez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Miguel A Olmedo-Suárez
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luis Ricardo Hernández-Barrientos
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Alejandro Alarcón-Del-Carmen
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Quetzalcoatl Escalante-Covarrubias
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lucía Mendoza-Viveros
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratorio de Cronobiología, Metabolismo y Envejecimiento, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), Mexico City, México
- Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, Mexico
| | - Enrique Hernández-Lemus
- Department of Computational Genomics, Centro de Ciencias de La Complejidad (C3), Instituto Nacional de Medicina Genómica (INMEGEN), Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfonso León-Del-Río
- Departamento de Medicina Genómica y Toxicología Ambiental, Programa Institucional de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Inti A de la Rosa-Velázquez
- Genomics Laboratory, Red de Apoyo a la Investigación-CIC, Universidad Nacional Autónoma de México, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080, Mexico City, Mexico
- Next Generation Sequencing Core Facility, Helmholtz Zentrum Muenchen, Ingolstaedter Landstr 1, 85754, Neuherberg, Germany
| | - Ricardo Orozco-Solis
- Laboratorio de Cronobiología, Metabolismo y Envejecimiento, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigación y de Estudios Avanzados (CIE-CINVESTAV), Mexico City, México
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
7
|
Rodríguez RM, Colom-Pellicer M, Hernández-Baixauli J, Calvo E, Suárez M, Arola-Arnal A, Torres-Fuentes C, Aragonès G, Mulero M. Grape Seed Proanthocyanidin Extract Attenuates Cafeteria-Diet-Induced Liver Metabolic Disturbances in Rats: Influence of Photoperiod. Int J Mol Sci 2024; 25:7713. [PMID: 39062955 PMCID: PMC11276873 DOI: 10.3390/ijms25147713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigated the influence of photoperiod (day length) on the efficacy of grape seed proanthocyanidin extract (GSPE) in mitigating metabolic disorders in obese rats fed a cafeteria diet. Rats were exposed to standard (L12), long (L18), or short (L6) photoperiods and treated with GSPE or vehicle. In the standard photoperiod, GSPE reduced body weight gain (50.5%), total cholesterol (37%), and triglycerides (34.8%), while increasing the expression of hepatic metabolic genes. In the long photoperiod, GSPE tended to decrease body weight gain, increased testosterone levels (68.3%), decreased liver weight (12.4%), and decreased reverse serum amino acids. In the short photoperiod, GSPE reduced glycemia (~10%) and lowered triglyceride levels (38.5%), with effects modified by diet. The standard photoperiod showed the greatest efficacy against metabolic syndrome-associated diseases. The study showed how day length affects GSPE's benefits and underscores considering biological rhythms in metabolic disease therapies.
Collapse
Affiliation(s)
- Romina M. Rodríguez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
| | - Marina Colom-Pellicer
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
| | - Julia Hernández-Baixauli
- Laboratory of Metabolism and Obesity, Vall d’Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Enrique Calvo
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Manuel Suárez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Anna Arola-Arnal
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Cristina Torres-Fuentes
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili (URV), Campus de Sescelades, 43007 Tarragona, Spain; (R.M.R.); (M.C.-P.); (E.C.); (M.S.); (A.A.-A.); (C.T.-F.); (G.A.)
- Center of Environmental, Food and Toxicological Technology-TecnATox, Rovira i Virgili University, 43201 Reus, Spain
| |
Collapse
|
8
|
Matsumura S, Signoretti C, Fatehi S, Tumenbayar BI, D'Addario C, Nimmer E, Thomas C, Viswanathan T, Wolf A, Garcia V, Rocic P, Bae Y, Alam SS, Gupte SA. Loss-of-function G6PD variant moderated high-fat diet-induced obesity, adipocyte hypertrophy, and fatty liver in male rats. J Biol Chem 2024; 300:107460. [PMID: 38876306 PMCID: PMC11328872 DOI: 10.1016/j.jbc.2024.107460] [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: 10/04/2023] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024] Open
Abstract
Obesity is a major risk factor for liver and cardiovascular diseases. However, obesity-driven mechanisms that contribute to the pathogenesis of multiple organ diseases are still obscure and treatment is inadequate. We hypothesized that increased , glucose-6-phosphate dehydrogenase (G6PD), the key rate-limiting enzyme in the pentose shunt, is critical in evoking metabolic reprogramming in multiple organs and is a significant contributor to the pathogenesis of liver and cardiovascular diseases. G6PD is induced by a carbohydrate-rich diet and insulin. Long-term (8 months) high-fat diet (HFD) feeding increased body weight and elicited metabolic reprogramming in visceral fat, liver, and aorta, of the wild-type rats. In addition, HFD increased inflammatory chemokines in visceral fat. Interestingly, CRISPR-edited loss-of-function Mediterranean G6PD variant (G6PDS188F) rats, which mimic human polymorphism, moderated HFD-induced weight gain and metabolic reprogramming in visceral fat, liver, and aorta. The G6PDS188F variant prevented HFD-induced CCL7 and adipocyte hypertrophy. Furthermore, the G6PDS188F variant increased Magel2 - a gene encoding circadian clock-related protein that suppresses obesity associated with Prader-Willi syndrome - and reduced HFD-induced non-alcoholic fatty liver. Additionally, the G6PDS188F variant reduced aging-induced aortic stiffening. Our findings suggest G6PD is a regulator of HFD-induced obesity, adipocyte hypertrophy, and fatty liver.
Collapse
Affiliation(s)
- Shun Matsumura
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | | | - Samuel Fatehi
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Bat Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Catherine D'Addario
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Erik Nimmer
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Colin Thomas
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Trisha Viswanathan
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Alexandra Wolf
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Petra Rocic
- Department of Physiology & Pharmacology, SHSU College of Osteopathic Medicine, Conroe, Texas, USA
| | - Yongho Bae
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Sm Shafiqul Alam
- Department of Pathology, Microbiology, and Immunology (PMI), New York Medical College, Valhalla, New York, USA
| | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA.
| |
Collapse
|
9
|
Su M, Yan Z, Wang Y, Cai J, Dong J, Luo J, Chen D, Liu A, Ye H. Depression Exacerbates Hepatic Steatosis in C57BL/6J Mice by Activating the Hypothalamic-Pituitary-Adrenal Axis. In Vivo 2024; 38:1677-1689. [PMID: 38936893 PMCID: PMC11215574 DOI: 10.21873/invivo.13618] [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: 02/10/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM Depression is associated with metabolic disorders, including non-alcoholic fatty liver disease (NAFLD). However, the mechanisms underlying the interaction between them are still poorly known. MATERIALS AND METHODS In this study, mice on a choline deficiency, L-amino acid-defined, high-fat diet (CDAHFD) developing steatosis were challenged with chronic restraint stress (CRS), a protocol widely used to induce depression. The development of depression and steatosis was evaluated using histopathology analysis, ELISA, q-PCR and Western Blot. RESULTS The contribution of the activated HPA axis to hepatic steatosis progress was fully established, which was validated using a hepatocyte model. Histopathological and biochemical analysis indicated that steatosis was exacerbated by CRS challenge, and behavioral tests indicated that the mice developed depression. Among the screened endocrinal pathways, the hypothalamic-pituitary-adrenal (HPA) axis was significantly activated and the synergistic effect of CDAHFD and CRS in activating the HPA axis was observed. In the hypothalamus, expression of corticotropin-releasing hormone (CRH) was increased by 86% and the protein levels of hypothalamic CRH were upregulated by 25% to 33% by CRS treatment. Plasma CRH levels were elevated by 45-56% and plasma adrenocorticotropic hormone (ACTH) levels were elevated by 29-58% by CRS treatment. In the liver, target genes of the HPA axis were activated, accompanied by disruption of the lipid metabolism and progression of steatohepatitis. The lipid metabolism in the Hepa1-6 cell line treated with endogenous corticosterone (CORT) was in accordance with the aforementioned in vivo responses. CONCLUSION Depression aggravated hepatic steatosis in CDAHFD-fed mice by activating the HPA axis. The risk of NAFLD development should be fully considered in depressive patients and improvement of psychotic disorders could be an etiological treatment strategy for them.
Collapse
Affiliation(s)
- Mingli Su
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Zheng Yan
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Ying Wang
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Jiacheng Cai
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Jia Dong
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Jia Luo
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Dahua Chen
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, P.R. China
| | - Aiming Liu
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Hua Ye
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, P.R. China;
| |
Collapse
|
10
|
Wu Y, Zhong M, Yin J, Ou W, Zhuang Y, Zhang N, Lin S, Zhu Y. A novel small-animal locomotor activity recording device for biological clock research. Animal Model Exp Med 2024; 7:71-76. [PMID: 38375555 PMCID: PMC10961864 DOI: 10.1002/ame2.12381] [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: 09/03/2023] [Accepted: 12/14/2023] [Indexed: 02/21/2024] Open
Abstract
The rodent running-wheel recording apparatus is a reliable approach for studying circadian rhythm. This study demonstrated how to construct a simple and intelligent running-wheel recording system. The running wheel was attached to the cage's base, whereas the Hall sensor was attached to the cage's cover. Then, the RJ25 adaptor relayed the running signal to the main control board. Finally, the main control board was connected to the USB port of the computer with the USB connection. Data were collected using the online-accessible, self-created software Magturning. Through Magturning, generated data were saved and exported in real time. Afterward, the device was validated by collecting data on the locomotor activities of mice under different light conditions. In conclusion, this new device can record circadian activity of rodents. Our device is appropriate for interdisciplinary investigations related to biological clock research.
Collapse
Affiliation(s)
- Yi‐Long Wu
- Endoscopy CenterThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Ming Zhong
- Department of EndocrinologyThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Jun Yin
- Chuangke Workshop Technology Co., LtdShenzhenChina
| | - Wei‐Jie Ou
- Department of Digestive NutritionFujian Children's HospitalFuzhouChina
| | - Yu‐Bin Zhuang
- Experimental Animal CenterFujian Medical UniversityFuzhouChina
| | - Nan‐Wen Zhang
- Department of PharmacologyFujian Medical UniversityFuzhouChina
| | - Su Lin
- Department of Hepatology, Hepatology Research Institute, Clinical Research Center for Liver and Intestinal Diseases of Fujian ProvinceThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| | - Yue‐Yong Zhu
- Department of Hepatology, Hepatology Research Institute, Clinical Research Center for Liver and Intestinal Diseases of Fujian ProvinceThe First Affiliated Hospital, Fujian Medical UniversityFuzhouChina
| |
Collapse
|
11
|
Schönberger K, Cabezas-Wallscheid N. How nutrition regulates hematopoietic stem cell features. Exp Hematol 2023; 128:10-18. [PMID: 37816445 DOI: 10.1016/j.exphem.2023.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023]
Abstract
Our dietary choices significantly impact all the cells in our body. Increasing evidence suggests that diet-derived metabolites influence hematopoietic stem cell (HSC) metabolism and function, thereby actively modulating blood homeostasis. This is of particular relevance because regulating the metabolic activity of HSCs is crucial for maintaining stem cell fitness and mitigating the risk of hematologic disorders. In this review, we examine the current scientific knowledge of the impact of diet on stemness features, and we specifically highlight the established mechanisms by which dietary components modulate metabolic and transcriptional programs in adult HSCs. Gaining a deeper understanding of how nutrition influences our HSC compartment may pave the way for targeted dietary interventions with the potential to decelerate aging and improve the effectiveness of transplantation and cancer therapies.
Collapse
|
12
|
He SK, Wang JH, Li T, Yin S, Cui JW, Xiao YF, Tang Y, Wang J, Bai YJ. Sleep and circadian rhythm disturbance in kidney stone disease: a narrative review. Front Endocrinol (Lausanne) 2023; 14:1293685. [PMID: 38089624 PMCID: PMC10711275 DOI: 10.3389/fendo.2023.1293685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
The circadian rhythm generated by circadian clock genes functions as an internal timing system. Since the circadian rhythm controls abundant physiological processes, the circadian rhythm evolved in organisms is salient for adaptation to environmental change. A disturbed circadian rhythm is a trigger for numerous pathological events. Recently, accumulated data have indicated that kidney stone disease (KSD) is related to circadian rhythm disturbance. However, the mechanism between them has not been fully elucidated. In this narrative review, we summarized existing evidence to illustrate the possible association between circadian rhythm disturbance and KSD based on the epidemiological studies and risk factors that are linked to circadian rhythm disturbance and discuss some chronotherapies for KSD. In summary, KSD is associated with systemic disorders. Metabolic syndrome, inflammatory bowel disease, and microbiome dysbiosis are the major risk factors supported by sufficient data to cause KSD in patients with circadian rhythm disturbance, while others including hypertension, vitamin D deficiency, parathyroid gland dysfunction, and renal tubular damage/dysfunction need further investigation. Then, some chronotherapies for KSD were confirmed to be effective, but the molecular mechanism is still unclear.
Collapse
Affiliation(s)
- Si-Ke He
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jia-Hao Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Li
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shan Yin
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jian-Wei Cui
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yun-Fei Xiao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yin Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jia Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yun-Jin Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Tuero C, Becerril S, Ezquerro S, Neira G, Frühbeck G, Rodríguez A. Molecular and cellular mechanisms underlying the hepatoprotective role of ghrelin against NAFLD progression. J Physiol Biochem 2023; 79:833-849. [PMID: 36417140 DOI: 10.1007/s13105-022-00933-1] [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: 09/06/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
The underlying mechanisms for the development and progression of nonalcoholic fatty liver disease (NAFLD) are complex and multifactorial. Within the last years, experimental and clinical evidences support the role of ghrelin in the development of NAFLD. Ghrelin is a gut hormone that plays a major role in the short-term regulation of appetite and long-term regulation of adiposity. The liver constitutes a target for ghrelin, where this gut-derived peptide triggers intracellular pathways regulating lipid metabolism, inflammation, and fibrosis. Interestingly, circulating ghrelin levels are altered in patients with metabolic diseases, such as obesity, type 2 diabetes, and metabolic syndrome, which, in turn, are well-known risk factors for the pathogenesis of NAFLD. This review summarizes the molecular and cellular mechanisms involved in the hepatoprotective action of ghrelin, including the reduction of hepatocyte lipotoxicity via autophagy and fatty acid β-oxidation, mitochondrial dysfunction, endoplasmic reticulum stress and programmed cell death, the reversibility of the proinflammatory phenotype in Kupffer cells, and the inactivation of hepatic stellate cells. Together, the metabolic and inflammatory pathways regulated by ghrelin in the liver support its potential as a therapeutic target to prevent NAFLD in patients with metabolic disorders.
Collapse
Affiliation(s)
- Carlota Tuero
- Department of General Surgery, Clínica Universidad de Navarra, School of Medicine, University of Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Silvia Ezquerro
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain.
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
14
|
Bolshette N, Ibrahim H, Reinke H, Asher G. Circadian regulation of liver function: from molecular mechanisms to disease pathophysiology. Nat Rev Gastroenterol Hepatol 2023; 20:695-707. [PMID: 37291279 DOI: 10.1038/s41575-023-00792-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2023] [Indexed: 06/10/2023]
Abstract
A wide variety of liver functions are regulated daily by the liver circadian clock and via systemic circadian control by other organs and cells within the gastrointestinal tract as well as the microbiome and immune cells. Disruption of the circadian system, as occurs during jetlag, shift work or an unhealthy lifestyle, is implicated in several liver-related pathologies, ranging from metabolic diseases such as obesity, type 2 diabetes mellitus and nonalcoholic fatty liver disease to liver malignancies such as hepatocellular carcinoma. In this Review, we cover the molecular, cellular and organismal aspects of various liver pathologies from a circadian viewpoint, and in particular how circadian dysregulation has a role in the development and progression of these diseases. Finally, we discuss therapeutic and lifestyle interventions that carry health benefits through support of a functional circadian clock that acts in synchrony with the environment.
Collapse
Affiliation(s)
- Nityanand Bolshette
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Hussam Ibrahim
- University of Düsseldorf, Medical Faculty, Institute of Clinical Chemistry and Laboratory Diagnostics, Düsseldorf, Germany
| | - Hans Reinke
- University of Düsseldorf, Medical Faculty, Institute of Clinical Chemistry and Laboratory Diagnostics, Düsseldorf, Germany.
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
15
|
Beckett LM, Donkin SS, Casey T. Circadian disruption decreases gluconeogenic flux in late-gestation, nonlactating dairy cows. JDS COMMUNICATIONS 2023; 4:412-416. [PMID: 37727248 PMCID: PMC10505771 DOI: 10.3168/jdsc.2022-0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/06/2023] [Indexed: 09/21/2023]
Abstract
Cattle exposed to shifts in light-dark phases during late pregnancy develop hypoglycemia and insulin resistance. Our objective was to investigate if differences in liver carbon flux for gluconeogenesis were driving circadian-disrupted metabolic alterations in glucose homeostasis, and relate changes in carbon flux to hepatic gene expression. We hypothesized circadian disruption would decrease hepatic carbon flux for glucose synthesis. Milking was ceased in late-gestation Holstein cows (n = 8) at 60 d before expected calving (BEC), and animals were assigned to either a control (n = 4) or a phase-shifted (PS; n = 4) group. From d 35 to 21 BEC both groups of cows were exposed to 16 h of light and 8 h of dark, but for the PS, light was shifted forward 6 h every 3 d. On d 21 BEC, liver biopsies were collected, subdivided, and incubated in 1.0 mM [U-13C] propionate for 2 h. Total RNA was isolated from a separate liver sample and used for RNA-sequencing analysis. Postincubation 13C mass isotopologue distribution was determined for aspartate, serine, alanine, and glutamate and used to calculate metabolic flux ratios. Enrichment of serine to enrichment of aspartate ratio (eSer:eAsp) was lower for PS (0.75 ± 0.02) cows compared with control (0.81 ± 0.04), indicating a reduction in carbon flux toward glucose for PS animals. eSer:eAsp ratio was negatively correlated to propionyl-CoA carboxylase (PCCB; r = -0.79) and succinate dehydrogenase subunit D (SDHD; r = -0.82). These relationships indicate that when dairy cattle are exposed to circadian disruption during late gestation, propionate carbon is preferentially used for energy rather than gluconeogenesis.
Collapse
Affiliation(s)
- Linda M. Beckett
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Shawn S. Donkin
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Theresa Casey
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
16
|
Deng G, Jiang Z, Lu H, Lu N, Zhu R, Zhu C, Zhou P, Tang X. A Study on the Amelioration of Circadian Rhythm Disorders in Fat Mice Using High-Protein Diets. Nutrients 2023; 15:3459. [PMID: 37571396 PMCID: PMC10421159 DOI: 10.3390/nu15153459] [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: 07/14/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
This innovative study investigates the effects of high-protein diets (milk protein) on the circadian rhythm of hepatic lipid metabolism. We aimed to understand how high-protein interventions regulate biological clock genes, maintain lipid metabolism balance, and affect the circadian rhythm of antioxidant levels in vivo. We divided 120 SPF-class C57BL/6J mice into the control, high-fat/low-protein (HF-LP), and high-fat/high-protein (HF-HP) groups. Mice were sacrificed during active (2 a.m. and 8 a.m.) and rest periods (2 p.m. and 8 p.m.). In the HF-LP group, hepatic lipid anabolic enzymes were consistently expressed at high levels, while key lipolytic enzymes slowly increased after feeding with no significant diurnal differences. This led to an abnormal elevation in blood lipid levels, a slow increase in and low levels of superoxide dismutase, and a rapid increase in malondialdehyde levels, deviating from the diurnal trend observed in the control group. However, high-protein interventions in the HF-HP group restored lipid synthase activity and the expression of key catabolic enzymes, exhibiting a precise circadian rhythm. It also improved the lipid-metabolism rhythm, which was disrupted by the high-fat diet. Overall, high-protein interventions restored the expression of key enzymes involved in lipid metabolism, improving the lipid-metabolism rhythm, which was disrupted by the high-fat diet.
Collapse
Affiliation(s)
- Guoliang Deng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
| | - Zhiqing Jiang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hui Lu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Rongxiang Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
| | - Chengkai Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
| | - Peng Zhou
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xue Tang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (G.D.); (Z.J.); (H.L.); (N.L.); (R.Z.); (C.Z.); (P.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
17
|
Hariri A, Mirian M, Zarrabi A, Kohandel M, Amini-Pozveh M, Aref AR, Tabatabaee A, Prabhakar PK, Sivakumar PM. The circadian rhythm: an influential soundtrack in the diabetes story. Front Endocrinol (Lausanne) 2023; 14:1156757. [PMID: 37441501 PMCID: PMC10333930 DOI: 10.3389/fendo.2023.1156757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/03/2023] [Indexed: 07/15/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) has been the main category of metabolic diseases in recent years due to changes in lifestyle and environmental conditions such as diet and physical activity. On the other hand, the circadian rhythm is one of the most significant biological pathways in humans and other mammals, which is affected by light, sleep, and human activity. However, this cycle is controlled via complicated cellular pathways with feedback loops. It is widely known that changes in the circadian rhythm can alter some metabolic pathways of body cells and could affect the treatment process, particularly for metabolic diseases like T2DM. The aim of this study is to explore the importance of the circadian rhythm in the occurrence of T2DM via reviewing the metabolic pathways involved, their relationship with the circadian rhythm from two perspectives, lifestyle and molecular pathways, and their effect on T2DM pathophysiology. These impacts have been demonstrated in a variety of studies and led to the development of approaches such as time-restricted feeding, chronotherapy (time-specific therapies), and circadian molecule stabilizers.
Collapse
Affiliation(s)
- Amirali Hariri
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Türkiye
| | - Mohammad Kohandel
- Department of Applied Mathematics, Faculty of Mathematics, University of Waterloo, Waterloo, ON, Canada
| | - Maryam Amini-Pozveh
- Department of Prosthodontics Dentistry, Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana Farber Cancer Institute, Boston, MA, United States
- Translational Sciences, Xsphera Biosciences Inc., Boston, MA, United States
| | - Aliye Tabatabaee
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pranav Kumar Prabhakar
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Division of Research and Development, Lovely Professional University, Phagwara Punjab, India
| | - Ponnurengam Malliappan Sivakumar
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
18
|
Jokl E, Llewellyn J, Simpson K, Adegboye O, Pritchett J, Zeef L, Donaldson I, Athwal VS, Purssell H, Street O, Bennett L, Guha IN, Hanley NA, Meng QJ, Piper Hanley K. Circadian Disruption Primes Myofibroblasts for Accelerated Activation as a Mechanism Underpinning Fibrotic Progression in Non-Alcoholic Fatty Liver Disease. Cells 2023; 12:1582. [PMID: 37371052 PMCID: PMC10297459 DOI: 10.3390/cells12121582] [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: 04/03/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/29/2023] Open
Abstract
Circadian rhythm governs many aspects of liver physiology and its disruption exacerbates chronic disease. CLOCKΔ19 mice disrupted circadian rhythm and spontaneously developed obesity and metabolic syndrome, a phenotype that parallels the progression of non-alcoholic fatty liver disease (NAFLD). NAFLD represents an increasing health burden with an estimated incidence of around 25% and is associated with an increased risk of progression towards inflammation, fibrosis and carcinomas. Excessive extracellular matrix deposition (fibrosis) is the key driver of chronic disease progression. However, little attention was paid to the impact of disrupted circadian rhythm in hepatic stellate cells (HSCs) which are the primary mediator of fibrotic ECM deposition. Here, we showed in vitro and in vivo that liver fibrosis is significantly increased when circadian rhythm is disrupted by CLOCK mutation. Quiescent HSCs from CLOCKΔ19 mice showed higher expression of RhoGDI pathway components and accelerated activation. Genes altered in this primed CLOCKΔ19 qHSC state may provide biomarkers for early liver disease detection, and include AOC3, which correlated with disease severity in patient serum samples. Integration of CLOCKΔ19 microarray data with ATAC-seq data from WT qHSCs suggested a potential CLOCK regulome promoting a quiescent state and downregulating genes involved in cell projection assembly. CLOCKΔ19 mice showed higher baseline COL1 deposition and significantly worse fibrotic injury after CCl4 treatment. Our data demonstrate that disruption to circadian rhythm primes HSCs towards an accelerated fibrotic response which worsens liver disease.
Collapse
Affiliation(s)
- Elliot Jokl
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Jessica Llewellyn
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Kara Simpson
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Oluwatobi Adegboye
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - James Pritchett
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK
| | - Leo Zeef
- Bioinformatics Core Facility, Faculty of Life Sciences, University of Manchester, Manchester M13 9PL, UK
| | - Ian Donaldson
- Bioinformatics Core Facility, Faculty of Life Sciences, University of Manchester, Manchester M13 9PL, UK
| | - Varinder S. Athwal
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Huw Purssell
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Oliver Street
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Lucy Bennett
- National Institute for Health Research, Nottingham Biomedical Research Centre, Nottingham University Hospitals National Health Service Trust, University of Nottingham, Nottingham NG7 2RD, UK
| | - Indra Neil Guha
- National Institute for Health Research, Nottingham Biomedical Research Centre, Nottingham University Hospitals National Health Service Trust, University of Nottingham, Nottingham NG7 2RD, UK
| | - Neil A. Hanley
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Qing-Jun Meng
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Karen Piper Hanley
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| |
Collapse
|
19
|
Ferreira V, Folgueira C, García-Altares M, Guillén M, Ruíz-Rosario M, DiNunzio G, Garcia-Martinez I, Alen R, Bookmeyer C, Jones JG, Cigudosa JC, López-Larrubia P, Correig-Blanchar X, Davis RJ, Sabio G, Rada P, Valverde ÁM. Hypothalamic JNK1-hepatic fatty acid synthase axis mediates a metabolic rewiring that prevents hepatic steatosis in male mice treated with olanzapine via intraperitoneal: Additional effects of PTP1B inhibition. Redox Biol 2023; 63:102741. [PMID: 37230004 DOI: 10.1016/j.redox.2023.102741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
Olanzapine (OLA), a widely used second-generation antipsychotic (SGA), causes weight gain and metabolic alterations when administered orally to patients. Recently, we demonstrated that, contrarily to the oral treatment which induces weight gain, OLA administered via intraperitoneal (i.p.) in male mice resulted in body weight loss. This protection was due to an increase in energy expenditure (EE) through a mechanism involving the modulation of hypothalamic AMPK activation by higher OLA levels reaching this brain region compared to those of the oral treatment. Since clinical studies have shown hepatic steatosis upon chronic treatment with OLA, herein we further investigated the role of the hypothalamus-liver interactome upon OLA administration in wild-type (WT) and protein tyrosine phosphatase 1B knockout (PTP1B-KO) mice, a preclinical model protected against metabolic syndrome. WT and PTP1B-KO male mice were fed an OLA-supplemented diet or treated via i.p. Mechanistically, we found that OLA i.p. treatment induces mild oxidative stress and inflammation in the hypothalamus in a JNK1-independent and dependent manner, respectively, without features of cell dead. Hypothalamic JNK activation up-regulated lipogenic gene expression in the liver though the vagus nerve. This effect concurred with an unexpected metabolic rewiring in the liver in which ATP depletion resulted in increased AMPK/ACC phosphorylation. This starvation-like signature prevented steatosis. By contrast, intrahepatic lipid accumulation was observed in WT mice treated orally with OLA; this effect being absent in PTP1B-KO mice. We also demonstrated an additional benefit of PTP1B inhibition against hypothalamic JNK activation, oxidative stress and inflammation induced by chronic OLA i.p. treatment, thereby preventing hepatic lipogenesis. The protection conferred by PTP1B deficiency against hepatic steatosis in the oral OLA treatment or against oxidative stress and neuroinflammation in the i.p. treatment strongly suggests that targeting PTP1B might be also a therapeutic strategy to prevent metabolic comorbidities in patients under OLA treatment in a personalized manner.
Collapse
Affiliation(s)
- Vitor Ferreira
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - María García-Altares
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain; Rovira I Virgili University, Department of Electronic Engineering, Tarragona, Spain
| | - Maria Guillén
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain
| | | | - Giada DiNunzio
- Center for Neurosciences and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Irma Garcia-Martinez
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Rosa Alen
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Christoph Bookmeyer
- Rovira I Virgili University, Department of Electronic Engineering, Tarragona, Spain
| | - John G Jones
- Center for Neurosciences and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | | | - Pilar López-Larrubia
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain
| | - Xavier Correig-Blanchar
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain; Rovira I Virgili University, Department of Electronic Engineering, Tarragona, Spain; Institut D'Investigacio Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Roger J Davis
- Program in Molecular Medicine, Chan Medical School, University of Massachusetts, Worcester, USA
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Patricia Rada
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain.
| | - Ángela M Valverde
- Instituto de Investigaciones Biomedicas Alberto Sols (IIBM), CSIC-UAM, Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain.
| |
Collapse
|
20
|
Areshidze DA, Kozlova MA, Mnikhovich MV, Bezuglova TV, Chernikov VP, Gioeva ZV, Borisov AV. Influence of Various Light Regimes on Morphofunctional Condition of Transplantable Melanoma B16. Biomedicines 2023; 11:biomedicines11041135. [PMID: 37189753 DOI: 10.3390/biomedicines11041135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
A study of the morphofunctional condition of mice with transplantable melanoma B16 under the influence of a normal daylight regime, constant lighting and constant darkness was conducted. It was shown that exposure to constant lighting leads to intensification of the proliferation of melanoma cells, more significant growth and spread of the tumor, the development of more pronounced secondary changes, the presence of perivascular growth and an increase in perineural invasion. At the same time, keeping of animals in constant darkness significantly reduced the intensity of the proliferative process in the tumor and lead to tumor regression in the absence of signs of lympho-, intravascular and intraneural invasion. Intergroup differences in tumor cell status were confirmed by the results of micromorphometric studies. It was also shown that the expression of clock genes was suppressed by an exposure to constant light, while an influence of constant darkness, on contrary, led to its intensification.
Collapse
Affiliation(s)
- David A Areshidze
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| | - Maria A Kozlova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| | - Maxim V Mnikhovich
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| | - Tatyana V Bezuglova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| | - Valery P Chernikov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| | - Zarina V Gioeva
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| | - Aleksey V Borisov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Center of Surgery", 117418 Moscow, Russia
| |
Collapse
|
21
|
Ribas-Aulinas F, Ribo S, Casas E, Mourin-Fernandez M, Ramon-Krauel M, Diaz R, Lerin C, Kalko SG, Vavouri T, Jimenez-Chillaron JC. Intergenerational Inheritance of Hepatic Steatosis in a Mouse Model of Childhood Obesity: Potential Involvement of Germ-Line microRNAs. Nutrients 2023; 15:nu15051241. [PMID: 36904241 PMCID: PMC10005268 DOI: 10.3390/nu15051241] [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: 01/10/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Childhood obesity increases the risk of developing metabolic syndrome later in life. Moreover, metabolic dysfunction may be inherited into the following generation through non-genomic mechanisms, with epigenetics as a plausible candidate. The pathways involved in the development of metabolic dysfunction across generations in the context of childhood obesity remain largely unexplored. We have developed a mouse model of early adiposity by reducing litter size at birth (small litter group, SL: 4 pups/dam; control group, C: 8 pups/dam). Mice raised in small litters (SL) developed obesity, insulin resistance and hepatic steatosis with aging. Strikingly, the offspring of SL males (SL-F1) also developed hepatic steatosis. Paternal transmission of an environmentally induced phenotype strongly suggests epigenetic inheritance. We analyzed the hepatic transcriptome in C-F1 and SL-F1 mice to identify pathways involved in the development of hepatic steatosis. We found that the circadian rhythm and lipid metabolic process were the ontologies with highest significance in the liver of SL-F1 mice. We explored whether DNA methylation and small non-coding RNAs might be involved in mediating intergenerational effects. Sperm DNA methylation was largely altered in SL mice. However, these changes did not correlate with the hepatic transcriptome. Next, we analyzed small non-coding RNA content in the testes of mice from the parental generation. Two miRNAs (miR-457 and miR-201) appeared differentially expressed in the testes of SL-F0 mice. They are known to be expressed in mature spermatozoa, but not in oocytes nor early embryos, and they may regulate the transcription of lipogenic genes, but not clock genes, in hepatocytes. Hence, they are strong candidates to mediate the inheritance of adult hepatic steatosis in our murine model. In conclusion, litter size reduction leads to intergenerational effects through non-genomic mechanisms. In our model, DNA methylation does not seem to play a role on the circadian rhythm nor lipid genes. However, at least two paternal miRNAs might influence the expression of a few lipid-related genes in the first-generation offspring, F1.
Collapse
Affiliation(s)
| | - Sílvia Ribo
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Eduard Casas
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | | | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana G. Kalko
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Tanya Vavouri
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- School of Medicine, University of Barcelona, L’Hospitalet, 08907 Barcelona, Spain
- Correspondence: or ; Tel.: +34-934024267
| |
Collapse
|
22
|
Down-regulation of hepatic CLOCK by PPARα is involved in inhibition of NAFLD. J Mol Med (Berl) 2023; 101:139-149. [PMID: 36527474 DOI: 10.1007/s00109-022-02279-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/24/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
This work aimed to investigate the role of nuclear factor peroxisome proliferator-activated receptor α (PPARα) in modification of circadian clock and their relevance to development of nonalcoholic fatty liver disease (NAFLD). Both male wild-type (WT) and Pparα-null (KO) mice treated with high-fat diet (HFD) were used to explore the effect of PPARα and lipid diet on the circadian rhythm. WT, KO, and PPARα-humanized (hPPARα) mice were treated with PPARα agonist fenofibrate to reveal the hPPARα dependence of circadian locomotor output cycles kaput (CLOCK) down-regulation. The mouse model and hepatocyte experiments were designed to verify the action of PPARα in down-regulating CLOCK and lipid accumulation in vivo and in vitro. Strongest NAFLD developed in mice fed 45%HFD, and it was inhibited in WT mice. The activity rhythm of WT mice was found to be different from that of the KO mice on normal diet and HFD. The core circadian factor CLOCK was down-regulated by HFD in both WT and KO mice in the liver, not in the hypothalamus. More interestingly, hepatic CLOCK was down-regulated by basal PPARα and activated PPARα in dose dependence of fenofibrate. Accordingly, CLOCK down-regulation dependent of PPARα activity was involved in inhibition of lipid metabolism in hepatocytes. Down-regulation of hepatic CLOCK by basal PPARα contributes to tolerance against development of NAFLD. Inhibition of CLOCK by activated PPARα is involved in inhibition of NAFLD by PPARα agonists. KEY MESSAGES: • PPARα inhibited NAFLD development induced by HFD. • PPARα mediated modifications of circadian rhythm and the hepatic circadian factor CLOCK in NAFLD models. • Down-regulation of hepatic CLOCK by basal PPARα contributed to tolerance against development of NAFLD. • Inhibition of CLOCK by activated PPARα was involved in therapeutic actions against fatty liver diseases by PPARα agonists.
Collapse
|
23
|
Circadian Rhythm Disorders Aggravate Periodontitis by Modulating BMAL1. Int J Mol Sci 2022; 24:ijms24010374. [PMID: 36613816 PMCID: PMC9820395 DOI: 10.3390/ijms24010374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Circadian rhythms regulate the body's homeostasis through the temporal control of tissue-specific circadian rhythm control genes. Circadian rhythm disorders (CRD) affect the expression levels of circadian rhythms-associated genes in brain and muscle aryl hydrocarbon receptor nuclear translocator-like-1(BMAL1), which is thought to contribute to metabolic disorders and an altered immune system. However, the relationship between CRD and the development of periodontitis was poorly reported. Therefore, this study aimed to investigate the role played by BMAL1 in periodontitis. We used a modified multi-platform approach (MMPM) to induce circadian rhythm disturbances in rats to investigate the role of BMAL1 in periodontitis. Our results showed significant downregulation of BMAL1 in the CRD with periodontitis group, significant resorption of alveolar bone, increased osteoclast differentiation, and upregulation of the inflammatory signaling molecule NF-κB. In addition, apoptosis and oxidative stress levels were increased in periodontal tissues. Collectively, our study suggests that BMAL1 is a key regulator in periodontitis exacerbated by CRD and that CRD may lead to the downregulation of BMAL1, thereby exacerbating oxidative stress and apoptosis in periodontal tissues. Our study found that BMAL1 may be associated with the progression of periodontitis and provides a new perspective on the treatment of periodontitis.
Collapse
|
24
|
Liu B, Wang S, Xu M, Ma Y, Sun R, Ding H, Li L. The double-edged role of hydrogen sulfide in the pathomechanism of multiple liver diseases. Front Pharmacol 2022; 13:899859. [PMID: 36588686 PMCID: PMC9800830 DOI: 10.3389/fphar.2022.899859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
In mammalian systems, hydrogen sulfide (H2S)-one of the three known gaseous signaling molecules in mammals-has been found to have a variety of physiological functions. Existing studies have demonstrated that endogenous H2S is produced through enzymatic and non-enzymatic pathways. The liver is the body's largest solid organ and is essential for H2S synthesis and elimination. Mounting evidence suggests H2S has essential roles in various aspects of liver physiological processes and pathological conditions, such as hepatic lipid metabolism, liver fibrosis, liver ischemia‒reperfusion injury, hepatocellular carcinoma, hepatotoxicity, and acute liver failure. In this review, we discuss the functions and underlying molecular mechanisms of H2S in multiple liver pathophysiological conditions.
Collapse
Affiliation(s)
- Bihan Liu
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Shanshan Wang
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China,Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ming Xu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yanan Ma
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Rui Sun
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Huiguo Ding
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lei Li
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China,*Correspondence: Lei Li,
| |
Collapse
|
25
|
Heintz MM, Eccles JA, Olack EM, Maner-Smith KM, Ortlund EA, Baldwin WS. Human CYP2B6 produces oxylipins from polyunsaturated fatty acids and reduces diet-induced obesity. PLoS One 2022; 17:e0277053. [PMID: 36520866 PMCID: PMC9754190 DOI: 10.1371/journal.pone.0277053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple factors in addition to over consumption lead to obesity and non-alcoholic fatty liver disease (NAFLD) in the United States and worldwide. CYP2B6 is the only human detoxification CYP whose loss is associated with obesity, and Cyp2b-null mice show greater diet-induced obesity with increased steatosis than wildtype mice. However, a putative mechanism has not been determined. LC-MS/MS revealed that CYP2B6 metabolizes PUFAs, with a preference for metabolism of ALA to 9-HOTrE and to a lesser extent 13-HOTrE with a preference for metabolism of PUFAs at the 9- and 13-positions. To further study the role of CYP2B6 in vivo, humanized-CYP2B6-transgenic (hCYP2B6-Tg) and Cyp2b-null mice were fed a 60% high-fat diet for 16 weeks. Compared to Cyp2b-null mice, hCYP2B6-Tg mice showed reduced weight gain and metabolic disease as measured by glucose tolerance tests, however hCYP2B6-Tg male mice showed increased liver triglycerides. Serum and liver oxylipin metabolite concentrations increased in male hCYP2B6-Tg mice, while only serum oxylipins increased in female hCYP2B6-Tg mice with the greatest increases in LA oxylipins metabolized at the 9 and 13-positions. Several of these oxylipins, specifically 9-HODE, 9-HOTrE, and 13-oxoODE, are PPAR agonists. RNA-seq data also demonstrated sexually dimorphic changes in gene expression related to nuclear receptor signaling, especially CAR > PPAR with qPCR suggesting PPARγ signaling is more likely than PPARα signaling in male mice. Overall, our data indicates that CYP2B6 is an anti-obesity enzyme, but probably to a lesser extent than murine Cyp2b's. Therefore, the inhibition of CYP2B6 by xenobiotics or dietary fats can exacerbate obesity and metabolic disease potentially through disrupted PUFA metabolism and the production of key lipid metabolites.
Collapse
Affiliation(s)
- Melissa M. Heintz
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Jazmine A. Eccles
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Emily M. Olack
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Kristal M. Maner-Smith
- Emory Integrated Metabolomics and Lipodomics Core, Emory University, Atlanta, Georgia, United States of America
| | - Eric A. Ortlund
- Department of Biochemistry, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
26
|
Perez-Diaz-del-Campo N, Castelnuovo G, Caviglia GP, Armandi A, Rosso C, Bugianesi E. Role of Circadian Clock on the Pathogenesis and Lifestyle Management in Non-Alcoholic Fatty Liver Disease. Nutrients 2022; 14:nu14235053. [PMID: 36501083 PMCID: PMC9736115 DOI: 10.3390/nu14235053] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Several features of the modern lifestyle, such as weekly schedules or irregular daily eating patterns, have become major drivers of global health problems, including non-alcoholic fatty liver disease (NAFLD). Sleep is an essential component of human well-being, and it has been observed that when circadian rhythms are disrupted, or when sleep quality decreases, an individual's overall health may worsen. In addition, the discrepancy between the circadian and social clock, due to weekly work/study schedules, is called social jetlag and has also been associated with adverse metabolic profiles. Current management of NAFLD is based on dietary intake and physical activity, with circadian preferences and other environmental factors also needing to be taken into account. In this regard, dietary approaches based on chrononutrition, such as intermittent fasting or time-restricted feeding, have proven to be useful in realigning lifestyle behaviors with circadian biological rhythms. However, more studies are needed to apply these dietary strategies in the treatment of these patients. In this review, we focus on the impact of circadian rhythms and the role of sleep patterns on the pathogenesis and development of NAFLD, as well as the consideration of chrononutrition for the precision nutrition management of patients with NAFLD.
Collapse
Affiliation(s)
| | | | | | - Angelo Armandi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Metabolic Liver Disease Research Program, I. Department of Medicine, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Chiara Rosso
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Correspondence:
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Gastroenterology Unit, Città della Salute e della Scienza—Molinette Hospital, 10126 Turin, Italy
| |
Collapse
|
27
|
Areshidze DA, Kozlova MA, Makartseva LA, Chernov IA, Sinelnikov MY, Kirillov YA. Influence of constant lightning on liver health: an experimental study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:83686-83697. [PMID: 35771326 DOI: 10.1007/s11356-022-21655-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
Light pollution has become a serious problem in many urbanized areas of the world. The impact of prolonged exposure to light and consequent disruption of natural circadian rhythms has significant health implications. The current study was undertaken to evaluate the effect of prolonged exposure to light, simulating urban light pollution, on liver health. In order to evaluate the effect of prolonged exposure to light, we examined the morphofunctional state, immunohistochemical and micromorphometric parameters of rat liver in normal conditions and following prolonged lighting exposure. Our results show that nocturnal light disruption triggers a cell death in the liver within 3 weeks (necrosis and apoptosis of hepatocytes) and stimulates a change in normal cellular karyometric parameters. At the same time, intracellular regeneration takes place within the organ, which manifests through hepatocyte hypertrophy. Under the influence of constant illumination, the circadian rhythms (CRs) of the size of hepatocytes and their nuclei are restructured, and the rhythm of the nuclear-cytoplasmic ratio is destroyed. The destruction of the CR of expression of p53 and Ki-67 also occurs against the background of the rearrangement of the daily rhythmicity of Per2 and Bmal1. The revealed changes in the morphofunctional state of the liver under the influence of light pollution indicate that a violation of normal illumination regimes is a potent factor leading to significant structural changes in the liver.
Collapse
Affiliation(s)
- David A Areshidze
- A.P. Avtsyn Research Institute of Human Morphology, Moscow, Russian Federation
| | - Maria A Kozlova
- A.P. Avtsyn Research Institute of Human Morphology, Moscow, Russian Federation
| | | | - Igor A Chernov
- Tyumen State Medical University, Tyumen, Russian Federation
| | - Mikhail Y Sinelnikov
- A.P. Avtsyn Research Institute of Human Morphology, Moscow, Russian Federation.
- Sechenov University, Moscow, Russian Federation.
| | - Yuri A Kirillov
- A.P. Avtsyn Research Institute of Human Morphology, Moscow, Russian Federation
| |
Collapse
|
28
|
Wang H, Zhang L, Xia Z, Cui JY. Effect of Chronic Cadmium Exposure on Brain and Liver Transporters and Drug-Metabolizing Enzymes in Male and Female Mice Genetically Predisposed to Alzheimer's Disease. Drug Metab Dispos 2022; 50:1414-1428. [PMID: 35878927 PMCID: PMC9513859 DOI: 10.1124/dmd.121.000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Cadmium (Cd) exposure is associated with increased Alzheimer's disease (AD) risks. The human Apolipoprotein E (ApoE) gene encodes a lipid-transporting protein that is critical for brain functions. Compared with ApoE2 and E3, ApoE4 is associated with increased AD risk. Xenobiotic biotransformation-related genes have been implicated in the pathogenesis of AD. However, little is known about the effects of Cd, ApoE, and sex on drug-processing genes. We investigated the Cd-ApoE interaction on the transcriptomic changes in the brains and livers of ApoE3/ApoE4 transgenic mice. Cd disrupts the transcriptomes of transporter and drug-processing genes in brain and liver in a sex- and ApoE-genotype-specific manner. Proinflammation related genes were enriched in livers of Cd-exposed ApoE4 males, whereas circadian rhythm and lipid metabolism related genes were enriched in livers of Cd-exposed ApoE3 females. In brains, Cd up-regulated the arachidonic acid-metabolizing Cyp2j isoforms only in the brains of ApoE3 mice, whereas the dysregulation of cation transporters was male-specific. In livers, several direct target genes of the major xenobiotic-sensing nuclear receptor pregnane X receptor were uniquely upregulated in Cd-exposed ApoE4 males. There was a female-specific hepatic upregulation of the steroid hormone-metabolizing Cyp2 isoforms and the bile acid synthetic enzyme Cyp7a1 by Cd exposure. The dysregulated liver transporters were mostly involved in intermediary metabolism, with the most significant response observed in ApoE3 females. In conclusion, Cd dysregulated the brain and liver drug-processing genes in a sex- and ApoE-genotype specific manner, and this may serve as a contributing factor for the variance in the susceptibility to Cd neurotoxicity. SIGNIFICANCE STATEMENT: Xenobiotic biotransformation plays an important role in modulating the toxicity of environmental pollutants. The human ApoE4 allele is the strongest genetic risk factor for AD, and cadmium (Cd) is increasingly recognized as an environmental factor of AD. Very little is known regarding the interactions between Cd exposure, sex, and the genes involved in xenobiotic biotransformation in brain and liver. The present study has addressed this critical knowledge gap.
Collapse
Affiliation(s)
- Hao Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Liang Zhang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Zhengui Xia
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| |
Collapse
|
29
|
Yu L, Lin C, Chen X, Teng Y, Zhou S, Liang Y. A Meta-Analysis of Sleep Disorders and Nonalcoholic Fatty Liver Disease: Potential Causality and Symptom Management. Gastroenterol Nurs 2022; 45:354-363. [PMID: 35913418 DOI: 10.1097/sga.0000000000000658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/29/2021] [Indexed: 01/10/2023] Open
Abstract
Nonalcoholic fatty liver disease is a type of metabolic disease, and recent research indicates that it may be associated with sleep disorders. We conducted a meta-analysis of current studies to estimate the associations between nonalcoholic fatty liver disease and sleep situation, including sleep duration, daytime sleepiness, and sleep disorder. This study follows the checklist of the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). Articles in the recent 10 years were searched from five databases. Eighteen articles, which met the eligibility criteria, were included in this meta-analysis. The results show that patients with nonalcoholic fatty liver disease have a shorter sleep duration and higher Epworth Sleepiness Scale score. Patients with short sleep duration (≤6 hours per night) or with obstructive sleep apnea have a higher risk of nonalcoholic fatty liver disease. In conclusion, there is a significant association between nonalcoholic fatty liver disease and sleep disorders in the included studies. In addition, patients with nonalcoholic fatty liver disease may have more severe daytime sleepiness and shorter sleep duration. More attention should be paid to the sleep situation of nonalcoholic fatty liver disease patients to potentially slow the disease progression.
Collapse
Affiliation(s)
- Lebing Yu
- Lebing Yu, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Chenli Lin, PhD, MD, is an associate professor, School of Medicine, Jinan University, Guangzhou, China
- Xinhe Chen, is BS MED candidate, School of Stomatology, Jinan University, Guangzhou, China
- Yuxin Teng, BSN, RN, is MPH candidate, School of Nursing, Jinan University, Guangzhou, China
- Shuang Zhou, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Yinji Liang, PhD, MD, RN, is an associate professor, School of Nursing, Jinan University, Guangzhou, China
| | - Chenli Lin
- Lebing Yu, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Chenli Lin, PhD, MD, is an associate professor, School of Medicine, Jinan University, Guangzhou, China
- Xinhe Chen, is BS MED candidate, School of Stomatology, Jinan University, Guangzhou, China
- Yuxin Teng, BSN, RN, is MPH candidate, School of Nursing, Jinan University, Guangzhou, China
- Shuang Zhou, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Yinji Liang, PhD, MD, RN, is an associate professor, School of Nursing, Jinan University, Guangzhou, China
| | - Xinhe Chen
- Lebing Yu, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Chenli Lin, PhD, MD, is an associate professor, School of Medicine, Jinan University, Guangzhou, China
- Xinhe Chen, is BS MED candidate, School of Stomatology, Jinan University, Guangzhou, China
- Yuxin Teng, BSN, RN, is MPH candidate, School of Nursing, Jinan University, Guangzhou, China
- Shuang Zhou, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Yinji Liang, PhD, MD, RN, is an associate professor, School of Nursing, Jinan University, Guangzhou, China
| | - Yuxin Teng
- Lebing Yu, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Chenli Lin, PhD, MD, is an associate professor, School of Medicine, Jinan University, Guangzhou, China
- Xinhe Chen, is BS MED candidate, School of Stomatology, Jinan University, Guangzhou, China
- Yuxin Teng, BSN, RN, is MPH candidate, School of Nursing, Jinan University, Guangzhou, China
- Shuang Zhou, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Yinji Liang, PhD, MD, RN, is an associate professor, School of Nursing, Jinan University, Guangzhou, China
| | - Shuang Zhou
- Lebing Yu, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Chenli Lin, PhD, MD, is an associate professor, School of Medicine, Jinan University, Guangzhou, China
- Xinhe Chen, is BS MED candidate, School of Stomatology, Jinan University, Guangzhou, China
- Yuxin Teng, BSN, RN, is MPH candidate, School of Nursing, Jinan University, Guangzhou, China
- Shuang Zhou, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Yinji Liang, PhD, MD, RN, is an associate professor, School of Nursing, Jinan University, Guangzhou, China
| | - Yinji Liang
- Lebing Yu, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Chenli Lin, PhD, MD, is an associate professor, School of Medicine, Jinan University, Guangzhou, China
- Xinhe Chen, is BS MED candidate, School of Stomatology, Jinan University, Guangzhou, China
- Yuxin Teng, BSN, RN, is MPH candidate, School of Nursing, Jinan University, Guangzhou, China
- Shuang Zhou, BSN, RN, is MSN candidate, School of Nursing, Jinan University, Guangzhou, China
- Yinji Liang, PhD, MD, RN, is an associate professor, School of Nursing, Jinan University, Guangzhou, China
| |
Collapse
|
30
|
Shen X, Zhang Y, Ji X, Li B, Wang Y, Huang Y, Zhang X, Yu J, Zou R, Qin D, Zhou H, Wang Q, Li JZ. Long Noncoding RNA lncRHL Regulates Hepatic VLDL Secretion by Modulating hnRNPU/BMAL1/MTTP Axis. Diabetes 2022; 71:1915-1928. [PMID: 35771993 PMCID: PMC9862400 DOI: 10.2337/db21-1145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/22/2022] [Indexed: 02/05/2023]
Abstract
Dysregulation of hepatic VLDL secretion contributes to the pathogenesis of metabolic diseases, such as nonalcoholic fatty liver disease (NAFLD) and hyperlipidemia. Accumulating evidence has suggested that long noncoding RNAs (lncRNAs) had malfunctioning roles in the pathogenesis of NAFLD. However, the function of lncRNAs in controlling hepatic VLDL secretion remains largely unillustrated. Here, we identified a novel lncRNA, lncRNA regulator of hyperlipidemia (lncRHL), which was liver-enriched, downregulated on high-fat diet feeding, and inhibited by oleic acid treatment in primary hepatocytes. With genetic manipulation in mice and primary hepatocytes, depletion of lncRHL induces hepatic VLDL secretion accompanied by decreased hepatic lipid contents. Conversely, lncRHL restoration reduces VLDL secretion with increased lipid deposition in hepatocytes. Mechanistic analyses indicate that lncRHL binds directly to heterogeneous nuclear ribonuclear protein U (hnRNPU), and thereby enhances its stability, and that hnRNPU can transcriptional activate Bmal1, leading to inhibition of VLDL secretion in hepatocytes. lncRHL deficiency accelerates the protein degradation of hnRNPU and suppresses the transcription of Bmal1, which in turn activates VLDL secretion in hepatocytes. With results taken together, we conclude that lncRHL is a novel suppressor of hepatic VLDL secretion. Activating the lncRHL/hnRNPU/BMAL1/MTTP axis represents a potential strategy for the maintenance of intrahepatic and plasma lipid homeostasis.
Collapse
Affiliation(s)
- Xuan Shen
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yajun Zhang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xuetao Ji
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Bo Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yuzhu Wang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yun Huang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xu Zhang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Jingxian Yu
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ruihan Zou
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Dongdong Qin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Wang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
- Shanghai Qi Zhi Institute, Shanghai, China
| |
Collapse
|
31
|
Fekry B, Ribas-Latre A, Drunen RV, Santos RB, Shivshankar S, Dai Y, Zhao Z, Yoo SH, Chen Z, Sun K, Sladek FM, Younes M, Eckel-Mahan K. Hepatic circadian and differentiation factors control liver susceptibility for fatty liver disease and tumorigenesis. FASEB J 2022; 36:e22482. [PMID: 35947136 PMCID: PMC10062014 DOI: 10.1096/fj.202101398r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 07/06/2022] [Accepted: 07/21/2022] [Indexed: 11/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer deaths, and the most common primary liver malignancy to present in the clinic. With the exception of liver transplant, treatment options for advanced HCC are limited, but improved tumor stratification could open the door to new treatment options. Previously, we demonstrated that the circadian regulator Aryl Hydrocarbon-Like Receptor Like 1 (ARNTL, or Bmal1) and the liver-enriched nuclear factor 4 alpha (HNF4α) are robustly co-expressed in healthy liver but incompatible in the context of HCC. Faulty circadian expression of HNF4α- either by isoform switching, or loss of expression- results in an increased risk for HCC, while BMAL1 gain-of-function in HNF4α-positive HCC results in apoptosis and tumor regression. We hypothesize that the transcriptional programs of HNF4α and BMAL1 are antagonistic in liver disease and HCC. Here, we study this antagonism by generating a mouse model with inducible loss of hepatic HNF4α and BMAL1 expression. The results reveal that simultaneous loss of HNF4α and BMAL1 is protective against fatty liver and HCC in carcinogen-induced liver injury and in the "STAM" model of liver disease. Furthermore, our results suggest that targeting Bmal1 expression in the absence of HNF4α inhibits HCC growth and progression. Specifically, pharmacological suppression of Bmal1 in HNF4α-deficient, BMAL1-positive HCC with REV-ERB agonist SR9009 impairs tumor cell proliferation and migration in a REV-ERB-dependent manner, while having no effect on healthy hepatocytes. Collectively, our results suggest that stratification of HCC based on HNF4α and BMAL1 expression may provide a new perspective on HCC properties and potential targeted therapeutics.
Collapse
Affiliation(s)
- Baharan Fekry
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Aleix Ribas-Latre
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Rachel Van Drunen
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Rafael Bravo Santos
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Samay Shivshankar
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, Texas, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, Texas, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, USA
| | - Seung-Hee Yoo
- Department of Biochemistry and Molecular Biology, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Zheng Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Kai Sun
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| | - Frances M Sladek
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, California, USA
| | - Mamoun Younes
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Kristin Eckel-Mahan
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA.,Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
32
|
Chen R, Ruan M, Chen S, Tian Y, Wang H, Li N, Zhang J, Yu X, Liu Z. Circadian dysregulation disrupts gut microbe-related bile acid metabolism. Food Nutr Res 2022. [DOI: 10.29219/fnr.v66.7653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Disturbance of circadian rhythm leads to abnormalities in bile acid (BA) and lipid metabolism, and it is of great significance to explore the relationship between them. This study explored the effects of circadian dysregulation on the rhythms of intestinal BA metabolism.
Method: Period circadian clock 1/period circadian clock 2 (Per1/Per2) double gene knockout (DKO) and wild-type (WT) male C57BL/6 mice were fed with a control or high-fat diet for 16 weeks. We measure plasma parameters of mice. Pathological changes including those in liver and intestine were detected by hematoxylin and eosin (H&E) and oil O staining. Western blot was used to detect the intestinal core rhythm protein clock circadian regulator (CLOCK), nuclear receptor subfamily 1, group D, member 1 (REV-ERBα), Farnesoid X receptor (FXR), Small heterodimer partner (SHP), and Fibroblast growth factor 15 (FGF15) expressions. We analyzed the bile acid and intestinal flora profile in the mice intestine tissues by BA-targeted metabolomics detection and high-throughput sequencing.
Results: Rhythmic chaos affected lipid metabolism and lipid accumulation in mice liver and intestine, and diurnal fluctuations of plasma triglycerides (TGs) were absent in normal-feeding DKO mice. The normal circadian fluctuations of the CLOCK and REV-ERBα observed in wild mice disappeared (normal diet) or were reversed (high-fat diet) in DKO mice. In WT mice intestine, total BA and conjugated BA were affected by circadian rhythm under both normal and high-fat diets, while these circadian fluctuations disappeared in DKO mice. Unconjugated BA seemed to be affected exclusively by diet (significantly increased in the high-fat group) without obvious fluctuations associated with circadian rhythm. Correlation analysis showed that the ratio of conjugated/unconjugated BA was positively correlated with the presence of Bacteroidetes and displayed a circadian rhythm. The expression levels of BA receptor pathway protein FXR, SHP, and FGF15 were affected by the ratio of conjugated/unconjugated BA.
Conclusion: Bacteroidetes-related diurnal changes to intestinal ratios of conjugated/unconjugated BA have the potential to regulate diurnal fluctuations in liver BA synthesis via FXR-FGF15. The inverted intestinal circadian rhythm observed in DKO mice fed with a high-fat diet may be an important reason for their abnormal circadian plasma TG rhythms and their susceptibility to lipid metabolism disorders.
Collapse
|
33
|
Aljabban J, Rohr M, Syed S, Khorfan K, Borkowski V, Aljabban H, Segal M, Mukhtar M, Mohammed M, Panahiazar M, Hadley D, Spengler R, Spengler E. Transcriptome changes in stages of non-alcoholic fatty liver disease. World J Hepatol 2022; 14:1382-1397. [PMID: 36158924 PMCID: PMC9376779 DOI: 10.4254/wjh.v14.i7.1382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/29/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the United States and globally. The currently understood model of pathogenesis consists of a ‘multiple hit’ hypothesis in which environmental and genetic factors contribute to hepatic inflammation and injury.
AIM To examine the genetic expression of NAFLD and non-alcoholic steatohepatitis (NASH) tissue samples to identify common pathways that contribute to NAFLD and NASH pathogenesis.
METHODS We employed the Search Tag Analyze Resource for Gene Expression Omnibus platform to search the The National Center for Biotechnology Information Gene Expression Omnibus to elucidate NAFLD and NASH pathology. For NAFLD, we conducted meta-analysis of data from 58 NAFLD liver biopsies and 60 healthy liver biopsies; for NASH, we analyzed 187 NASH liver biopsies and 154 healthy liver biopsies.
RESULTS Our results from the NAFLD analysis reinforce the role of altered metabolism, inflammation, and cell survival in pathogenesis and support recently described contributors to disease activity, such as altered androgen and long non-coding RNA activity. The top upstream regulator was found to be sterol regulatory element binding transcription factor 1 (SREBF1), a transcription factor involved in lipid homeostasis. Downstream of SREBF1, we observed upregulation in CXCL10, HMGCR, HMGCS1, fatty acid binding protein 5, paternally expressed imprinted gene 10, and downregulation of sex hormone-binding globulin and insulin-like growth factor 1. These molecular changes reflect low-grade inflammation secondary to accumulation of fatty acids in the liver. Our results from the NASH analysis emphasized the role of cholesterol in pathogenesis. Top canonical pathways, disease networks, and disease functions were related to cholesterol synthesis, lipid metabolism, adipogenesis, and metabolic disease. Top upstream regulators included pro-inflammatory cytokines tumor necrosis factor and IL1B, PDGF BB, and beta-estradiol. Inhibition of beta-estradiol was shown to be related to derangement of several cellular downstream processes including metabolism, extracellular matrix deposition, and tumor suppression. Lastly, we found riciribine (an AKT inhibitor) and ZSTK-474 (a PI3K inhibitor) as potential drugs that targeted the differential gene expression in our dataset.
CONCLUSION In this study we describe several molecular processes that may correlate with NAFLD disease and progression. We also identified ricirbine and ZSTK-474 as potential therapy.
Collapse
Affiliation(s)
- Jihad Aljabban
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Michael Rohr
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, United States
| | - Saad Syed
- Department of Medicine, Northwestern Memorial Hospital, Chicago, IL 60611, United States
| | - Kamal Khorfan
- Department of Gastroenterology and Hepatology, University of California San Francisco-Fresno , Fresno, CA 93701, United States
| | - Vincent Borkowski
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Hisham Aljabban
- Department of Medicine, Barry University, Miami, FL 33161, United States
| | - Michael Segal
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Mohamed Mukhtar
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI 49503, United States
| | - Mohammed Mohammed
- Department of Medicine, Windsor University School of Medicine, Saint Kitts 1621, Cayon, Saint Kitts and Nevis
| | - Maryam Panahiazar
- Department of Surgery, University of California San Francisco, San Francisco, CA 94305, United States
| | - Dexter Hadley
- Department of Artificial Intelligence, Pathology, University of Central Florida College of Medicine , Orlando, FL 32827, United States
| | - Ryan Spengler
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Erin Spengler
- Department of Gastroenterology and Hepatology, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| |
Collapse
|
34
|
Skrlec I, Talapko J. Hepatitis B and circadian rhythm of the liver. World J Gastroenterol 2022; 28:3282-3296. [PMID: 36158265 PMCID: PMC9346465 DOI: 10.3748/wjg.v28.i27.3282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/15/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm in humans is determined by the central clock located in the hypothalamus’s suprachiasmatic nucleus, and it synchronizes the peripheral clocks in other tissues. Circadian clock genes and clock-controlled genes exist in almost all cell types. They have an essential role in many physiological processes, including lipid metabolism in the liver, regulation of the immune system, and the severity of infections. In addition, circadian rhythm genes can stimulate the immune response of host cells to virus infection. Hepatitis B virus (HBV) infection is the leading cause of liver disease and liver cancer globally. HBV infection depends on the host cell, and hepatocyte circadian rhythm genes are associated with HBV replication, survival, and spread. The core circadian rhythm proteins, REV-ERB and brain and muscle ARNTL-like protein 1, have a crucial role in HBV replication in hepatocytes. In addition to influencing the virus’s life cycle, the circadian rhythm also affects the pharmacokinetics and efficacy of antiviral vaccines. Therefore, it is vital to apply antiviral therapy at the appropriate time of day to reduce toxicity and improve the effectiveness of antiviral treatment. For these reasons, understanding the role of the circadian rhythm in the regulation of HBV infection and host responses to the virus provides us with a new perspective of the interplay of the circadian rhythm and anti-HBV therapy. Therefore, this review emphasizes the importance of the circadian rhythm in HBV infection and the optimization of antiviral treatment based on the circadian rhythm-dependent immune response.
Collapse
Affiliation(s)
- Ivana Skrlec
- Department of Biophysics, Biology, and Chemistry, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Jasminka Talapko
- Department of Anatomy Histology, Embryology, Pathology Anatomy and Pathology Histology, Faculty of Dental Medicine and Health, Osijek 31000, Croatia
| |
Collapse
|
35
|
Johnson H, Yates T, Leedom G, Ramanathan C, Puppa M, van der Merwe M, Tipirneni-Sajja A. Multi-Tissue Time-Domain NMR Metabolomics Investigation of Time-Restricted Feeding in Male and Female Nile Grass Rats. Metabolites 2022; 12:metabo12070657. [PMID: 35888782 PMCID: PMC9321200 DOI: 10.3390/metabo12070657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/06/2022] [Accepted: 07/13/2022] [Indexed: 02/05/2023] Open
Abstract
Metabolic disease resulting from overnutrition is prevalent and rapidly increasing in incidence in modern society. Time restricted feeding (TRF) dietary regimens have recently shown promise in attenuating some of the negative metabolic effects associated with chronic nutrient stress. The purpose of this study is to utilize a multi-tissue metabolomics approach using nuclear magnetic resonance (NMR) spectroscopy to investigate TRF and sex-specific effects of high-fat diet in a diurnal Nile grass rat model. Animals followed a six-week dietary protocol on one of four diets: chow ad libitum, high-fat ad libitum (HF-AD), high-fat early TRF (HF-AM), or high-fat late TRF (HF-PM), and their liver, heart, and white adipose tissues were harvested at the end of the study and were analyzed by NMR. Time-domain complete reduction to amplitude–frequency table (CRAFT) was used to semi-automate and systematically quantify metabolites in liver, heart, and adipose tissues while minimizing operator bias. Metabolite profiling and statistical analysis revealed lipid remodeling in all three tissues and ectopic accumulation of cardiac and hepatic lipids for HF-AD feeding compared to a standard chow diet. Animals on TRF high-fat diet had lower lipid levels in the heart and liver compared to the ad libitum group; however, no significant differences were noted for adipose tissue. Regardless of diet, females exhibited greater amounts of hepatic lipids compared to males, while no consistent differences were shown in adipose and heart. In conclusion, this study demonstrates the feasibility of performing systematic and time-efficient multi-tissue NMR metabolomics to elucidate metabolites involved in the crosstalk between different metabolic tissues and provides a more holistic approach to better understand the etiology of metabolic disease and the effects of TRF on metabolic profiles.
Collapse
Affiliation(s)
- Hayden Johnson
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA; (H.J.); (T.Y.); (G.L.)
| | - Thomas Yates
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA; (H.J.); (T.Y.); (G.L.)
| | - Gary Leedom
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA; (H.J.); (T.Y.); (G.L.)
| | - Chidambaram Ramanathan
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (C.R.); (M.P.); (M.v.d.M.)
| | - Melissa Puppa
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (C.R.); (M.P.); (M.v.d.M.)
| | - Marie van der Merwe
- College of Health Sciences, University of Memphis, Memphis, TN 38152, USA; (C.R.); (M.P.); (M.v.d.M.)
| | - Aaryani Tipirneni-Sajja
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA; (H.J.); (T.Y.); (G.L.)
- Correspondence:
| |
Collapse
|
36
|
Imre L, Niaki EF, Bosire R, Nanasi P, Nagy P, Bacso Z, Hamidova N, Pommier Y, Jordan A, Szabo G. Nucleosome destabilization by polyamines. Arch Biochem Biophys 2022; 722:109184. [PMID: 35395253 PMCID: PMC10572104 DOI: 10.1016/j.abb.2022.109184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 11/19/2022]
Abstract
The roles and molecular interactions of polyamines (PAs) in the nucleus are not fully understood. Here their effect on nucleosome stability, a key regulatory factor in eukaryotic gene control, is reported, as measured in agarose embedded nuclei of H2B-GFP expressor HeLa cells. Nucleosome stability was assessed by quantitative microscopy [1,2] in situ, in close to native state of chromatin, preserving the nucleosome constrained topology of the genomic DNA. A robust destabilizing effect was observed in the millimolar concentration range in the case of spermine, spermidine as well as putrescine, which was strongly pH and salt concentration-dependent, and remained significant also at neutral pH. The integrity of genomic DNA was not affected by PA treatment, excluding DNA break-elicited topological relaxation as a factor in destabilization. The binding of PAs to DNA was demonstrated by the displacement of ethidium bromide, both from deproteinized nuclear halos and from plasmid DNA. The possibility that DNA methylation patterns may be influenced by PA levels is contemplated in the context of gene expression and DNA methylation correlations identified in the NCI-60 panel-based CellMiner database: methylated loci in subsets of high-ODC1 cell lines and the dependence of PER3 DNA methylation on PA metabolism.
Collapse
Affiliation(s)
- Laszlo Imre
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Erfaneh Firouzi Niaki
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Rosevalentine Bosire
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Peter Nanasi
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Peter Nagy
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Nubar Hamidova
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary
| | - Yves Pommier
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4255, USA
| | - Albert Jordan
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, 08028, Spain
| | - Gabor Szabo
- Department of Biophysics and Cell Biology, University of Debrecen, Faculty of Medicine Debrecen, H-4032, Hungary.
| |
Collapse
|
37
|
Apple polyphenol extract targets circadian rhythms to improve liver biological clock and lipid homeostasis in C57BL/6 male mice with mistimed high-fat diet feeding. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
38
|
Xiao P, Goodarzi P, Pezeshki A, Hagen DE. RNA-seq reveals insights into molecular mechanisms of metabolic restoration via tryptophan supplementation in low birth weight piglet model. J Anim Sci 2022; 100:skac156. [PMID: 35552417 PMCID: PMC9155244 DOI: 10.1093/jas/skac156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
Low birth weight (LBW) is associated with metabolic disorders in early life. While dietary l-tryptophan (Trp) can ameliorate postprandial plasma triglycerides (TG) disposal in LBW piglets, the genetic and biological basis underlying Trp-caused alterations in lipid metabolism is poorly understood. In this study, we collected 24 liver samples from 1-mo-old LBW and normal birth weight (NBW) piglets supplemented with different concentrations of dietary Trp (NBW with 0% Trp, N0; LBW with 0% Trp, L0; LBW with 0.4% Trp, L4; LBW with 0.8% Trp, L8; N = 6 in each group.) and conducted systematic, transcriptome-wide analysis using RNA sequencing (RNA-seq). We identified 39 differentially expressed genes (DEG) between N0 and L0, and genes within "increased dose effect" clusters based on dose-series expression profile analysis, enriched in fatty acid response of gene ontology (GO) biological process (BP). We then identified RNA-binding proteins including SRSF1, DAZAP1, PUM2, PCBP3, IGF2BP2, and IGF2BP3 significantly (P < 0.05) enriched in alternative splicing events (ASE) in comparison with L0 as control. There were significant positive and negative relationships between candidate genes from co-expression networks (including PID1, ANKRD44, RUSC1, and CYP2J34) and postprandial plasma TG concentration. Further, we determined whether these candidate hub genes were also significantly associated with metabolic and cardiovascular traits in humans via human phenome-wide association study (Phe-WAS), and analysis of mammalian orthologs suggests a functional conservation between human and pig. Our work demonstrates that transcriptomic changes during dietary Trp supplementation in LBW piglets. We detected candidate genes and related BP that may play roles on lipid metabolism restoration. These findings will help to better understand the amino acid support in LBW metabolic complications.
Collapse
Affiliation(s)
- Ping Xiao
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Parniyan Goodarzi
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Adel Pezeshki
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Darren E Hagen
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
39
|
Soliz-Rueda JR, López-Fernández-Sobrino R, Bravo FI, Aragonès G, Suarez M, Muguerza B. Grape Seed Proanthocyanidins Mitigate the Disturbances Caused by an Abrupt Photoperiod Change in Healthy and Obese Rats. Nutrients 2022; 14:nu14091834. [PMID: 35565801 PMCID: PMC9100649 DOI: 10.3390/nu14091834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Variations in the light/dark cycle and obesogenic diets trigger physiological and behavioral disorders. Proanthocyanidins, in addition to their healthy properties, have recently demonstrated a modulating effect on biological rhythms. Therefore, the aim of this study was to evaluate the administration of a grape seed proanthocyanidin-rich extract (GSPE) to mitigate the disruption caused by a sudden photoperiod change in healthy and cafeteria (CAF)-diet obese rats. For this, 48 photoperiod-sensitive Fischer 344 rats were fed standard or CAF diets for 6 weeks under a standard (12 h light/day, L12) conditions. Then, rats were switched to a long (18 h light/day, L18) or short (6 h light/day, L6) photoperiod and administered vehicle or GSPE (25 mg/kg) for 1 week. Body weight (BW) and food intake (FI) were recorded weekly. Animal activity and serum hormone concentrations were studied before and after the photoperiod change. Hormone levels were measured both at 3 h (ZT3) and 15 h (ZT15) after the onset of light. Results showed the impact of the CAF diet and photoperiod on the BW, FI, activity, and hormonal status of the animals. GSPE administration resulted in an attenuation of the changes produced by the photoperiod disruption. Specifically, GSPE in L6 CAF-fed rats reduced serum corticosterone concentration, restoring its circadian rhythm, increased the T3-to-T4 ratio, and increased light phase activity, while under L18, it decreased BW and testosterone concentration and increased the animal activity. These results suggest that GSPE may contribute to the adaptation to the new photoperiods. However, further studies are needed to elucidate the metabolic pathways and processes involved in these events.
Collapse
|
40
|
Schroder EA, Delisle BP. Time Restricted Feeding to the Light Cycle Dissociates Canonical Circadian Clocks and Physiological Rhythms in Heart Rate. Front Pharmacol 2022; 13:910195. [PMID: 35645828 PMCID: PMC9133719 DOI: 10.3389/fphar.2022.910195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Circadian rhythms are approximate 24-h biological cycles that optimize molecular and physiological functions to predictable daily environmental changes in order to maintain internal and organismal homeostasis. Environmental stimuli (light, feeding, activity) capable of altering the phase of molecular rhythms are important tools employed by circadian biologists to increase understanding of the synchronization of circadian rhythms to the environment and to each other within multicellular systems. The central circadian clock, located in the suprachiasmatic nucleus (SCN) of the hypothalamus is largely responsive to light and is thought to entrain the phase of peripheral clocks via neurohumoral signals. Mice are nocturnal and consume most of their food during the dark cycle. Early studies demonstrated that altered metabolic cues in the form of time restricted feeding, specifically, feeding mice during the light cycle, resulted in an uncoupling of molecular clocks in peripheral tissues with those from the SCN. These studies showed as much as a 12-h shift in gene expression in some peripheral tissues but not others. The shifts occurred without corresponding changes in the central clock in the brain. More recent studies have demonstrated that changes in cardiac physiology (heart rate, MAP) in response to time of food intake occur independent of the cardiac molecular clock. Understanding differences in the physiology/function and gene expression in other organs both independently and in relation to the heart in response to altered feeding will be important in dissecting the roles of the various clocks throughout the body, as well as, understanding their links to cardiovascular pathology.
Collapse
Affiliation(s)
- Elizabeth A. Schroder
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kentucky, Lexington, KY, United States
- *Correspondence: Elizabeth A. Schroder, ; Brian P. Delisle,
| | - Brian P. Delisle
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- *Correspondence: Elizabeth A. Schroder, ; Brian P. Delisle,
| |
Collapse
|
41
|
Kozlova MA, Kirillov YA, Makartseva LA, Chernov I, Areshidze DA. Morphofunctional State and Circadian Rhythms of the Liver under the Influence of Chronic Alcohol Intoxication and Constant Lighting. Int J Mol Sci 2021; 22:ijms222313007. [PMID: 34884810 PMCID: PMC8657715 DOI: 10.3390/ijms222313007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 01/10/2023] Open
Abstract
A study of the influence of chronic alcohol intoxication, constant illumination and their combined effects on the morphofunctional state of the rat liver and the circadian rhythms (CR) of the studied parameters of the organism was carried out. It was found that both alcohol and constant illumination caused significant changes in the structure of the liver, as well as in the circadian rhythmicity of micromorphometric parameters of hepatocytes, ALT, and total and direct bilirubin rhythms; however, the combined effects of ethanol and constant illumination had the most significant effect on the studied parameters of the organism. These two factors caused disturbances in the circadian rhythms of the micromorphometric parameters of hepatocytes, disruption of the circadian rhythms of total protein, albumin, AST, ALT, and direct and total bilirubin, as well as disturbances in the expression and rhythmicity of the studied clock genes against a background of the development of an inflammatory process in the liver.
Collapse
Affiliation(s)
- Maria A. Kozlova
- Laboratory of Cell Pathology, A.P. Avtsyn Research Institute of Human Morphology, 117218 Moscow, Russia; (M.A.K.); (Y.A.K.); (L.A.M.)
| | - Yuri A. Kirillov
- Laboratory of Cell Pathology, A.P. Avtsyn Research Institute of Human Morphology, 117218 Moscow, Russia; (M.A.K.); (Y.A.K.); (L.A.M.)
| | - Lyudmila A. Makartseva
- Laboratory of Cell Pathology, A.P. Avtsyn Research Institute of Human Morphology, 117218 Moscow, Russia; (M.A.K.); (Y.A.K.); (L.A.M.)
| | - Igor Chernov
- Department of Pathological Anatomy, Tyumen State Medical University, 625023 Tyumen, Russia;
| | - David A. Areshidze
- Laboratory of Cell Pathology, A.P. Avtsyn Research Institute of Human Morphology, 117218 Moscow, Russia; (M.A.K.); (Y.A.K.); (L.A.M.)
- Experimental Tumor Chemotherapy Group, Center for Screening and Preclinical Testing, Institute of Problems of Chemical Physics of the Russian Academy of Science, 142432 Chernogolovka, Russia
- Correspondence: ; Tel.: +7-909-643-37-56
| |
Collapse
|
42
|
Feng C, Liu W, Chen H, Dong W, Yang J. Effect of dark environment on intestinal flora and expression of genes related to liver metabolism in zebrafish (Danio rerio). Comp Biochem Physiol C Toxicol Pharmacol 2021; 249:109100. [PMID: 34174412 DOI: 10.1016/j.cbpc.2021.109100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/19/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023]
Abstract
To explore the effects of dark environment on intestinal flora and expression of genes related to liver metabolism in zebrafish, a total of 60 zebrafish were fed for 21 days (24 h dark treatments or 14/10 h light/dark cycle), and the influence of dark environment on gut microbes and liver gene expression was studied using sequencing analysis of intestinal flora and liver. The results showed that the body weight of fish was significantly increased in the dark group than that in the control group (P < 0.05). Compared with the control group, dark environment treatment changed the composition of dominant flora, increased the abundance of unconventional bacteria and reduced probiotics in the intestine of zebrafish. Of these, the ratio of Bacteroidetes to Firmicutes in the intestine was reduced. The genome expression of the liver showed significant changes, and liver metabolites were also affected. Meanwhile, dark environment decreased gene expression associated with changes in blood glucose, lipid metabolism and immunization. Dark environment also caused liver steatosis as observed by histological study. This study shows that dark environment treatment has an important impact on liver metabolism and intestinal microbes in zebrafish.
Collapse
Affiliation(s)
- Chi Feng
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China
| | - Wuyun Liu
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China; School of Animal Science, Mongolian State University of Agriculture, Bayangol, Ulaanbaatar, Mongolia
| | - Hao Chen
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China
| | - Wu Dong
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China
| | - Jingfeng Yang
- Inner Mongolia Key Laboratory of Toxicant Monitoring and Toxicology, College of Animal Science and Technology, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia 028000, China.
| |
Collapse
|
43
|
Ribas-Aulinas F, Parra-Vargas M, Ramon-Krauel M, Diaz R, Lerin C, Cambras T, Jimenez-Chillaron JC. Time-Restricted Feeding during Puberty Ameliorates Adiposity and Prevents Hepatic Steatosis in a Mouse Model of Childhood Obesity. Nutrients 2021; 13:3579. [PMID: 34684586 PMCID: PMC8538558 DOI: 10.3390/nu13103579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Time restricted feeding (TRF) refers to dietary interventions in which food access is limited during a specific timeframe of the day. TRFs have proven useful in improving metabolic health in adult subjects with obesity. Their beneficial effects are mediated, in part, through modulating the circadian rhythm. Nevertheless, the translation of these dietary interventions onto obese/overweight children and adolescents remains uncharacterized. The objective of this study is to explore the feasibility of temporal dietary interventions for improving metabolic health in the context of childhood obesity. METHODS We have previously developed a mouse model of early adiposity (i.e., childhood obesity) through litter size reduction. Mice raised in small litters (SL) became obese as early as by two weeks of age, and as adults, they developed several obesity-related co-morbidities, including insulin resistance, glucose intolerance and hepatic steatosis. Here, we explored whether two independent short-term chrono-nutritional interventions might improve metabolic health in 1-month-old pre-pubertal SL mice. Both TRFs comprised 8 h feeding/14 h fasting. In the first one (TRF1) Control and SL mice had access to the diet for 8 h during the dark phase. In the second intervention (TRF2) food was available during the light:dark transitions. RESULTS TRF1 did not alter food intake nor ameliorate adiposity in SL-TRF1. In contrast, SL-TRF2 mice showed unintentional reduction of caloric intake, which was accompanied by reduced total body weight and adiposity. Strikingly, hepatic triglyceride content was completely normalized in SL-TRF1 and SL-TRF2 mice, when compared to the ad lib-fed SL mice. These effects were partially mediated by (i) clock-dependent signals, which might modulate the expression of Pparg or Cpt1a, and (ii) clock-independent signals, such as fasting itself, which could influence Fasn expression. CONCLUSIONS Time-restricted feeding is an effective and feasible nutritional intervention to improve metabolic health, namely hepatic steatosis, in a model of childhood obesity. These data open new avenues for future safe and efficient chrono-nutritional interventions aimed to improve metabolic health in children with overweight/obesity.
Collapse
Affiliation(s)
- Francesc Ribas-Aulinas
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| | - Marcela Parra-Vargas
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
- School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
- School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| | - Trinitat Cambras
- Department of Biochemistry and Physiology, School of Pharmacy, University of Barcelona, 08028 Barcelona, Spain;
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain; (F.R.-A.); (M.P.-V.); (M.R.-K.); (R.D.); (C.L.)
| |
Collapse
|
44
|
Litwinowicz K, Waszczuk E, Gamian A. Advanced Glycation End-Products in Common Non-Infectious Liver Diseases: Systematic Review and Meta-Analysis. Nutrients 2021; 13:3370. [PMID: 34684371 PMCID: PMC8537188 DOI: 10.3390/nu13103370] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Excessive intake of fructose, glucose and alcohol is associated with the development of non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). At the same time, these dietetic factors create an environment favorable for the generation of advanced glycation end-products. For this reason, advanced glycation end-products (AGEs) are hypothesized to play role in the development of NAFLD and ALD. In this systematic review and meta-analysis, we explore the relationship between NAFLD and ALD with AGE levels, including their diagnostic accuracy. METHODS The systematic review and meta-analysis has been pre-registered with PROSPERO (CRD42021240954) and was performed in accordance with the PRISMA guidelines. Meta-analyses were performed using the meta R package. RESULTS We have obtained 11 studies meeting our inclusion criteria, reporting data on 1844 participants (909 with NAFLD, 169 with ALD and 766 healthy controls). NAFLD was associated with significantly higher AGE fluorescence and serum N-(carboxyethyl)lysine (CEL) levels. Patients with alcoholic cirrhosis had significantly higher levels of N-(carboxymethyl)lysine (CML). Only individual studies examined AGEs in the context of their diagnostic accuracy. AGE fluorescence distinguished low and moderate steatosis with an AUC of 0.76. The ratio of CML, CEL and pentosidine to a soluble variant of the AGE receptor differentiated patients with NAFLD from healthy controls with high AUC (0.83-0.85). Glyceraldehyde-derived AGE separated non-alcoholic fatty liver (NAFL) from non-alcoholic steatohepatitis (NASH) with acceptable performance (AUC 0.78). CONCLUSIONS In conclusion, NAFLD and ALD are associated with significantly higher levels of several AGEs. More research is needed to examine the diagnostic accuracy of AGEs, however individual studies show that AGEs perform well in distinguishing NAFL from NASH.
Collapse
Affiliation(s)
- Kamil Litwinowicz
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wrocław, Poland
| | - Ewa Waszczuk
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, 50-556 Wrocław, Poland;
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland;
| |
Collapse
|
45
|
Ribas-Aulinas F, Ribo S, Parra-Vargas M, Fernández-Pérez A, Cebrià J, Guardiola-Perello M, Ramon-Krauel M, Lerin C, Diaz R, Kalko SG, Vallejo M, Díez-Noguera A, Cambras T, Jimenez-Chillaron JC. Neonatal overfeeding during lactation rapidly and permanently misaligns the hepatic circadian rhythm and programmes adult NAFLD. Mol Metab 2021; 45:101162. [PMID: 33422644 PMCID: PMC7851182 DOI: 10.1016/j.molmet.2021.101162] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 01/20/2023] Open
Abstract
Childhood obesity is a strong risk factor for adult obesity, type 2 diabetes, and cardiovascular disease. The mechanisms that link early adiposity with late-onset chronic diseases are poorly characterised. We developed a mouse model of early adiposity through litter size reduction. Mice reared in small litters (SLs) developed obesity, insulin resistance, and hepatic steatosis during adulthood. The liver played a major role in the development of the disease. OBJECTIVE To gain insight into the molecular mechanisms that link early development and childhood obesity with adult hepatic steatosis and insulin resistance. METHODS We analysed the hepatic transcriptome (Affymetrix) of control and SL mice to uncover potential pathways involved in the long-term programming of disease in our model. RESULTS The circadian rhythm was the most significantly deregulated Gene Ontology term in the liver of adult SL mice. Several core clock genes, such as period 1-3 and cryptochrome 1-2, were altered in two-week-old SL mice and remained altered throughout their life course until they reached 4-6 months of age. Defective circadian rhythm was restricted to the periphery since the expression of clock genes in the hypothalamus, the central pacemaker, was normal. The period-cryptochrome genes were primarily entrained by dietary signals. Hence, restricting food availability during the light cycle only uncoupled the central rhythm from the peripheral and completely normalised hepatic triglyceride content in adult SL mice. This effect was accompanied by better re-alignment of the hepatic period genes, suggesting that they might have played a causal role in mediating hepatic steatosis in the adult SL mice. Functional downregulation of Per2 in hepatocytes in vitro confirmed that the period genes regulated lipid-related genes in part through peroxisome proliferator-activated receptor alpha (Ppara). CONCLUSIONS The hepatic circadian rhythm matures during early development, from birth to postnatal day 30. Hence, nutritional challenges during early life may misalign the hepatic circadian rhythm and secondarily lead to metabolic derangements. Specific time-restricted feeding interventions improve metabolic health in the context of childhood obesity by partially re-aligning the peripheral circadian rhythm.
Collapse
Affiliation(s)
- Francesc Ribas-Aulinas
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain
| | - Silvia Ribo
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain
| | - Marcela Parra-Vargas
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain
| | - Antonio Fernández-Pérez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid y Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Madrid, Spain
| | - Judith Cebrià
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain
| | - Maria Guardiola-Perello
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain
| | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain; Departament de Medicina, Facultat de Medicina, Universitat de Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain; Departament de Medicina, Facultat de Medicina, Universitat de Barcelona, Spain
| | | | - Mario Vallejo
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid y Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas CIBERDEM, Madrid, Spain
| | - Antoni Díez-Noguera
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Spain
| | - Trinitat Cambras
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Spain
| | - Josep C Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu (Saint John of God Children's Hospital Barcelona), Endocrinology, Esplugues, Barcelona, Spain.
| |
Collapse
|
46
|
Unraveling the Role of Leptin in Liver Function and Its Relationship with Liver Diseases. Int J Mol Sci 2020; 21:ijms21249368. [PMID: 33316927 PMCID: PMC7764544 DOI: 10.3390/ijms21249368] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/19/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Since its discovery twenty-five years ago, the fat-derived hormone leptin has provided a revolutionary framework for studying the physiological role of adipose tissue as an endocrine organ. Leptin exerts pleiotropic effects on many metabolic pathways and is tightly connected with the liver, the major player in systemic metabolism. As a consequence, understanding the metabolic and hormonal interplay between the liver and adipose tissue could provide us with new therapeutic targets for some chronic liver diseases, an increasing problem worldwide. In this review, we assess relevant literature regarding the main metabolic effects of leptin on the liver, by direct regulation or through the central nervous system (CNS). We draw special attention to the contribution of leptin to the non-alcoholic fatty liver disease (NAFLD) pathogenesis and its progression to more advanced stages of the disease as non-alcoholic steatohepatitis (NASH). Likewise, we describe the contribution of leptin to the liver regeneration process after partial hepatectomy, the mainstay of treatment for certain hepatic malignant tumors.
Collapse
|
47
|
Abstract
Circadian rhythms are biological systems that synchronize cellular circadian oscillators with the organism's daily feeding-fasting or rest-activity cycles in mammals. Circadian rhythms regulate nutrient absorption and utilization at the cellular level and are closely related to obesity and metabolic disorders. Bile acids are important modulators that facilitate nutrient absorption and regulate energy metabolism. Here, we provide an overview of the current connections and future perspectives between the circadian clock and bile acid metabolism as well as related metabolic diseases. Feeding and fasting cycles influence bile acid pool size and composition, and bile acid signaling can respond to acute lipid and glucose utilization and mediate energy balance. Disruption of circadian rhythms such as shift work, irregular diet, and gene mutations can contribute to altered bile acid metabolism and heighten obesity risk. High-fat diets, alcohol, and gene mutations related to bile acid signaling result in desynchronized circadian rhythms. Gut microbiome also plays a role in connecting circadian rhythms with bile acid metabolism. The underlying mechanism of how circadian rhythms interact with bile acid metabolism has not been fully explored. Sustaining bile acid homeostasis based on circadian rhythms may be a potential therapy to alleviate metabolic disturbance.
Collapse
Affiliation(s)
- Yunxia Yang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, China
| |
Collapse
|
48
|
Fujita SI, Rutter L, Ong Q, Muratani M. Integrated RNA-seq Analysis Indicates Asynchrony in Clock Genes between Tissues under Spaceflight. Life (Basel) 2020; 10:E196. [PMID: 32933026 PMCID: PMC7555136 DOI: 10.3390/life10090196] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Rodent models have been widely used as analogs for estimating spaceflight-relevant molecular mechanisms in human tissues. NASA GeneLab provides access to numerous spaceflight omics datasets that can potentially generate novel insights and hypotheses about fundamental space biology when analyzed in new and integrated fashions. Here, we performed a pilot study to elucidate space biological mechanisms across tissues by reanalyzing mouse RNA-sequencing spaceflight data archived on NASA GeneLab. Our results showed that clock gene expressions in spaceflight mice were altered compared with those in ground control mice. Furthermore, the results suggested that spaceflight promotes asynchrony of clock gene expressions between peripheral tissues. Abnormal circadian rhythms are associated not only with jet lag and sleep disorders but also with cancer, lifestyle-related diseases, and mental disorders. Overall, our findings highlight the importance of elucidating the causes of circadian rhythm disruptions using the unique approach of space biology research to one day potentially develop countermeasures that benefit humans on Earth and in space.
Collapse
Affiliation(s)
- Shin-Ichiro Fujita
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Lindsay Rutter
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Quang Ong
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
49
|
Zhang H, Dahlén T, Khan A, Edgren G, Rzhetsky A. Measurable health effects associated with the daylight saving time shift. PLoS Comput Biol 2020; 16:e1007927. [PMID: 32511231 PMCID: PMC7302868 DOI: 10.1371/journal.pcbi.1007927] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/18/2020] [Accepted: 05/06/2020] [Indexed: 12/26/2022] Open
Abstract
The transition to daylight saving time (DST) is beneficial for energy conservation but at the same time it has been reported to increase the risk of cerebrovascular and cardiovascular problems. Here, we evaluate the effect of the DST shift on a whole spectrum of diseases-an analysis we hope will be helpful in weighing the risks and benefits of DST shifts. Our study relied on a population-based, cross-sectional analysis of the IBM Watson Health MarketScan insurance claim dataset, which incorporates over 150 million unique patients in the US, and the Swedish national inpatient register, which incorporates more than nine million unique Swedes. For hundreds of sex- and age-specific diseases, we assessed effects of the DST shifts forward and backward by one hour in spring and autumn by comparing the observed and expected diagnosis rates after DST shift exposure. We found four prominent, elevated risk clusters, including cardiovascular diseases (such as heart attacks), injuries, mental and behavioral disorders, and immune-related diseases such as noninfective enteritis and colitis to be significantly associated with DST shifts in the United States and Sweden. While the majority of disease risk elevations are modest (a few percent), a considerable number of diseases exhibit an approximately ten percent relative risk increase. We estimate that each spring DST shift is associated with negative health effects-with 150,000 incidences in the US, and 880,000 globally. We also identify for the first time a collection of diseases with relative risks that appear to decrease immediately after the spring DST shift, enriched with infections and immune system-related maladies. These diseases' decreasing relative risks might be driven by the documented boosting effect of a short-term stress (such as that experienced around the spring DST shift) on the immune system.
Collapse
Affiliation(s)
- Hanxin Zhang
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, Illinois, United States of America
- Department of Medicine, and Institute of Genomics and Systems Biology, The University of Chicago, Chicago, Illinois, United States of America
| | - Torsten Dahlén
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Atif Khan
- Department of Medicine, and Institute of Genomics and Systems Biology, The University of Chicago, Chicago, Illinois, United States of America
| | - Gustaf Edgren
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Södersjukhuset Hospital, Stockholm, Sweden
| | - Andrey Rzhetsky
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, Illinois, United States of America
- Department of Medicine, and Institute of Genomics and Systems Biology, The University of Chicago, Chicago, Illinois, United States of America
- Department of Human Genetics and Committee on Quantitative Methods in Social, Behavioral, and Health Sciences, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
50
|
Sun HJ, Wu ZY, Nie XW, Wang XY, Bian JS. Implications of hydrogen sulfide in liver pathophysiology: Mechanistic insights and therapeutic potential. J Adv Res 2020; 27:127-135. [PMID: 33318872 PMCID: PMC7728580 DOI: 10.1016/j.jare.2020.05.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Background Over the last several decades, hydrogen sulfide (H2S) has been found to exert multiple physiological functions in mammal systems. The endogenous production of H2S is primarily mediated by cystathione β-synthase (CBS), cystathione γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3-MST). These enzymes are widely expressed in the liver tissues and regulate hepatic functions by acting on various molecular targets. Aim of Review In the present review, we will highlight the recent advancements in the cellular events triggered by H2S under liver diseases. The therapeutic effects of H2S donors on hepatic diseases will also be discussed. Key Scientific Concepts of Review As a critical regulator of liver functions, H2S is critically involved in the etiology of various liver disorders, such as nonalcoholic steatohepatitis (NASH), hepatic fibrosis, hepatic ischemia/reperfusion (IR) injury, and liver cancer. Targeting H2S-producing enzymes may be a promising strategy for managing hepatic disorders.
Collapse
Key Words
- 3-MP, 3-mercaptopyruvate
- 3-MST, 3-mercaptopyruvate sulfurtransferase
- AGTR1, angiotensin II type 1 receptor
- AMPK, AMP-activated protein kinase
- Akt, protein kinase B
- CAT, cysteine aminotransferase
- CBS, cystathione β-synthase
- CO, carbon monoxide
- COX-2, cyclooxygenase-2
- CSE, cystathione γ-lyase
- CX3CR1, chemokine CX3C motif receptor 1
- Cancer
- DAO, D-amino acid oxidase
- DATS, Diallyl trisulfide
- EGFR, epidermal growth factor receptor
- ERK, extracellular regulated protein kinases
- FAS, fatty acid synthase
- Fibrosis
- H2S, hydrogen sulfide
- HFD, high fat diet
- HO-1, heme oxygenase 1
- Hydrogen sulfide
- IR, ischemia/reperfusion
- Liver disease
- MMP-2, matrix metalloproteinase 2
- NADH, nicotinamide adenine dinucleotide
- NADPH, nicotinamide adenine dinucleotide phosphate
- NAFLD, non-alcoholic fatty liver diseases
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-kappa B
- NaHS, sodium hydrosulfide
- Nrf2, nuclear factor erythroid2-related factor 2
- PI3K, phosphatidylinositol 3-kinase
- PLP, pyridoxal 5′-phosphate
- PPG, propargylglycine
- PTEN, phosphatase and tensin homolog deleted on chromosome ten
- SAC, S-allyl-cysteine
- SPRC, S-propargyl-cysteine
- STAT3, signal transducer and activator of transcription 3
- Steatosis
- VLDL, very low density lipoprotein
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Zhi-Yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Xiao-Wei Nie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Xin-Yu Wang
- Department of Endocrinology, The First Affiliated Hospital of Shenzhen University (Shenzhen Second People's Hospital), Shenzhen 518037, China
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore.,National University of Singapore Research Institute, Suzhou 215000, China
| |
Collapse
|