1
|
Lakhani HA, Biswas D, Kuruvila M, Chava MS, Raj K, Varghese JT, Swathi NL. Intermittent fasting versus continuous caloric restriction for glycemic control and weight loss in type 2 diabetes: A traditional review. Prim Care Diabetes 2025; 19:203-213. [PMID: 40000314 DOI: 10.1016/j.pcd.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
Type 2 Diabetes Mellitus (T2DM) represents a major global health issue, with its incidence anticipated to increase markedly in the forthcoming decades. Efficient non-pharmacological therapies, especially dietary approaches, are essential for regulating glycemic control and facilitating weight reduction. Intermittent Fasting (IF) and Continuous Caloric Restriction (CCR) are two well researched methodologies, but their relative effectiveness and enduring advantages continue to be topics of active discussion. This review systematically assesses and contrasts the impacts of intermittent fasting (IF) and continuous calorie restriction (CCR) on glycemic regulation and weight reduction in persons with type 2 diabetes mellitus (T2DM), highlighting their short-term and long-term effects, safety profiles, and adherence rates. A thorough literature analysis was performed utilizing PubMed and Google Scholar, concentrating on papers published from 2000 to 2024. The review encompassed randomized controlled trials and observational studies that investigated the effects of intermittent fasting (IF) and continuous calorie restriction (CCR) on glycemic indicators (HbA1c, fasting glucose) and body weight. IF shown substantial short-term advantages, encompassing marked decreases in HbA1c levels, fasting glucose, and body weight. Mechanistic discoveries emphasized better insulin sensitivity, augmented fat metabolism, and autophagy as key aspects. In contrast, CCR was linked to enduring metabolic enhancements, including decreased visceral fat and improved insulin sensitivity. Nevertheless, both dietary approaches demonstrated constraints.
Collapse
Affiliation(s)
- Hairya Ajaykumar Lakhani
- Internal medicine, Smt. B.K. Shah Medical Institute and Research Centre, Sumandeep Vidyapeeth, Waghodia Road, Vadodara, Gujarat 391760, India.
| | - Deepanwita Biswas
- Internal medicine, Bharati Vidyapeeth Deemed to be University Medical College and Hospital, Bharati Vidyapeeth, Dhankawadi, Pune, Maharashtra 411043, India
| | - Mahima Kuruvila
- Internal medicine, Caribbean Medical University School of Medicine, 5600 N River Rd #800, Rosemont, IL 60018, USA
| | - Manisha Sai Chava
- Internal medicine, Kakatiya Medical College, Rangampet street, Warangal, Telangana 506007, India
| | - Kshitij Raj
- Internal medicine, Grant Government Medical College, Mumbai, India
| | - Joel Thomas Varghese
- Internal Medicine, Rak medical and health sciences university, Ras Al Khaimah, UAE
| | - N L Swathi
- Pharm D, Department of Pharmacy Practice, Jawaharlal Nehru Technological University, Anantapuramu, Anantapur, Andhra Pradesh, India
| |
Collapse
|
2
|
Li T, Chen G, Cao L, Rong W, Zhao H, Xiong Z, Liu Q, Song J, Wang W, Liu Y, Wang X, Liu S. Effects of combined exposure to 17α-methyltestosterone and polystyrene microplastics on lipid metabolism and the nervous system in Danio rerio. J Steroid Biochem Mol Biol 2025; 247:106665. [PMID: 39694074 DOI: 10.1016/j.jsbmb.2024.106665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/20/2024]
Abstract
Polystyrene (PS) microplastics are pervasive environmental pollutants that are harmful to aquatic organisms upon degradation. The synthetic androgen 17α-methyltestosterone (MT) is an environmental endocrine-disrupting chemical. This study aimed to systematically evaluate the combined histological and molecular effects of MT and PS exposure on the liver and brain tissues of Danio rerio with focus on lipid metabolism and neural function disruption. Female D. rerio were exposed to 50 ng/L MT and 0.5 mg/L PS (5 μm in diameter) for 21 d. Histological observations, real-time quantitative PCR (qPCR), and RNA-sequencing (RNA-seq) analysis were employed to assess the effects of PS and MT. These results indicated that MT and PS co-exposure caused fatty degeneration of liver cells and a significant upregulation of lipid synthesis-related genes (ACSS1, CEL, FASN, and GK5). In brain tissue, the observed effects included reduced marginal layer neuron counts, cytoplasmic loosening of central layer neurons, disordered gray matter layer cells, and vascular congestion. RNA-seq analysis further revealed significant enrichment of differentially expressed genes in the "glycine, serine, and threonine metabolism" and "neuroactive ligand-receptor interaction" signaling pathways. Thus, MT and PS co-exposure induced lipid metabolism disorders in D. rerio and influence neural signaling by altering the "neuroactive ligand-receptor interaction" pathway. These findings highlight the complex risks posed by environmental pollutants to aquatic life and provide critical insights for environmental protection and aquatic health research.
Collapse
Affiliation(s)
- Tongyao Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Gen Chen
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Lu Cao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Weiya Rong
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Haiyan Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Zijun Xiong
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Qing Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Jing Song
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Weiwei Wang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Yu Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xianzong Wang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China.
| | - Shaozhen Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China.
| |
Collapse
|
3
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [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: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
4
|
Qin Z, Chen J, Liu F, Li B, Zhang C, Wang X, Liu L, Wang M, Wang T, Wang S, Yu F, Wang S, Yang J. Jellyfish stings-induced cardiac failure was ameliorated through AAG-mediated glycogen-driven ATP production. EXPLORATION (BEIJING, CHINA) 2025; 5:20230089. [PMID: 40040825 PMCID: PMC11875447 DOI: 10.1002/exp.20230089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/16/2024] [Indexed: 07/27/2024]
Abstract
Jellyfish stings have become a common injury among fishermen and divers. Severe jellyfish stings could worsen cardiac function and even cause cardiac complications, ultimately leading to cardiac failure (CF). Currently, there are no effective drugs available. Single cell sequencing revealed alpha-1 acid glycoprotein (AAG), an energy regulatory protein targeting to glycogen, was highly expressed in jellyfish stings-induced CF patients. However, the mechanism remains elusive. It is postulated that AAG could increase glycogen metabolism, protecting against jellyfish stings-induced CF. AAG deletion exacerbated CF, while exogenous and endogenous AAG ameliorated CF. AAG also rescued the decline triggered by the AAG knockout (KO). Intriguingly, AAG improved cardiac function and metabolic adaptation by glycogen-driven ATP production, shifting mitochondrial/glycolytic ATP production towards glycolysis. Sorted by single-cell RNA sequencing and spatial transcription technology, CC-chemokine receptor 5 (CCR5) and Peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1α) were differentially expressed. Mechanistically, CCR5 inhibitor MVC abolished AAG's protective effect and PGC-1α overexpression. Collectively, jellyfish stings-induced CF was ameliorated through AAG-mediated glycogen-driven ATP production, promoting glycolytic/mitochondrial metabolic switches to rely energetically primarily on glycolysis, which might serve as a therapeutic target of CF.
Collapse
Affiliation(s)
- Zhen Qin
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jinhong Chen
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Fang Liu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Bingbing Li
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Chenchen Zhang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Xiuxiu Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Lin Liu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
- School of Health Sciences and EngineeringUniversity of Shanghai for Science and TechnologyShanghaiChina
| | - Mingke Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | | | - Su Wang
- Center for Translational NeuromedicineUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Feifei Yu
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Shifeng Wang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| | - Jishun Yang
- PLA Naval Medical CenterNaval Medical University (Second Military Medical University)ShanghaiChina
| |
Collapse
|
5
|
Guo H, Sun Y, Gao W, Liu Y, Han T, Zhang D. The association of activity patterns on female reproductive diseases: a prospective cohort study of UK biobank. BMC Public Health 2025; 25:312. [PMID: 39856643 PMCID: PMC11762564 DOI: 10.1186/s12889-025-21576-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] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Little is known about the role of timing of physical activity in female reproductive disorders. These disorders include polycystic ovary syndrome (PCOS), heavy menstrual bleeding (HMB), endometriosis, infertility, and pregnancy-related disorders. This study aims to investigate the associations of activity patterns with female reproductive diseases. METHODS A total of 49,540 female participants from the UK Biobank with valid accelerometer data were enrolled at baseline. Activity patterns were defined based on the timing of moderate-to-vigorous intensity physical activity (MVPA) throughout the day. Participants were categorized into four groups according to the timing of their MVPA: "morning, evening, mixed, midday-afternoon", with the midday-afternoon group serving as the reference. Cox proportional hazards models were utilized to evaluate the association between activity patterns and female reproductive diseases. RESULTS During a median follow-up of 12.6 years, a total of 1044 cases of female reproductive diseases were documented. After adjustment for potential confounders, compared to women with midday-afternoon exercise, women with morning exercise and mixed-timing exercise were associated with lower risks for female reproductive diseases (HRmorning=0.81, 95% CI: 0.67-0.98; HRmixed=0.79, 95% CI: 0.69-0.91, P-trend < 0.05). Moreover, morning exercise and mixed-timing exercise had lower risks of PCOS (HRmorning=0.38, 95% CI: 0.15-0.97; HRmixed=0.27, 95% CI: 0.13-0.57, P-trend<0.001), and mixed-timing exercise was associated with a lower risk for HMB (HRmixed=0.81, 95% CI: 0.70-0.95, P-trend < 0.05), compared with the reference group. CONCLUSIONS Compared with midday-afternoon group, morning and mixed MVPA timing groups, but not evening group, were associated with decreased risks for female reproductive diseases and PCOS. In addition, we found that women with mixed MVPA timing exercise had a lower risk of HMB, compared with the reference group.
Collapse
Affiliation(s)
- Huanyu Guo
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Harbin, 150081, Heilongjiang, P. R. China
| | - Yingzi Sun
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Harbin, 150081, Heilongjiang, P. R. China
| | - Wei Gao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Harbin, 150081, Heilongjiang, P. R. China
| | - Yuqing Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Harbin, 150081, Heilongjiang, P. R. China
| | - Tianshu Han
- National Key Discipline Laboratory, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China.
| | - Dandan Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Harbin, 150081, Heilongjiang, P. R. China.
| |
Collapse
|
6
|
Dutta P, Annoor A, Dey P, Sultana J, Mokbul MI, Naurin SA, Roy R, Simona SY, Dutta J, Mazumder T, Masud F. The Link Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Gastroesophageal Reflux Disease. Cureus 2024; 16:e71095. [PMID: 39512967 PMCID: PMC11542734 DOI: 10.7759/cureus.71095] [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] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly nonalcoholic fatty liver disease (NAFLD), and gastroesophageal reflux disease (GERD) are prevalent chronic conditions with escalating global incidence. This study delves into the intricate interplay between MASLD and GERD. The primary objective is to comprehensively explore the association between MASLD and GERD, investigating how various factors contribute to the coexistence and potential exacerbation of these conditions. We conducted a literature search in PubMed and Google Scholar of only human studies over the past 10 years. The search included systematic review, meta-analysis, editorial, and cross-sectional studies of patients with MASLD and GERD. The prevalence of GERD in patients with MASLD was higher, with various risk factors coming into play. Obesity was identified as an independent risk factor for both GERD and MASLD. However, obese patients predominantly had higher disease progression. Lifestyle factors like physical activity and dietary modifications emerge as promising strategies to mitigate risk.
Collapse
Affiliation(s)
| | | | - Proma Dey
- Internal Medicine, Chittagong Medical College, Chattagram, BGD
| | - Jakia Sultana
- Internal Medicine, Comilla Medical College, Cumilla, BGD
| | | | | | - Ritwik Roy
- Internal Medicine, Ragib-Rabeya Medical College and Hospital, Chittagong, BGD
| | | | - Jui Dutta
- Internal Medicine, Comilla Medical College, Cumilla, BGD
| | - Tanusree Mazumder
- Family Medicine, Zainul Haque Sikder Women's Medical College and Hospital, Dhaka, BGD
| | - Farjana Masud
- Internal Medicine, Shaheed Ziaur Rahman Medical College, Dhaka, BGD
| |
Collapse
|
7
|
Ohbayashi K, Sugiyama Y, Nohmi T, Nishimura K, Nakazaki T, Sato YI, Masumura T, Iwasaki Y. Anekomochi glutinous rice provides low postprandial glycemic response by enhanced insulin action via GLP-1 release and vagal afferents activation. J Physiol Sci 2024; 74:47. [PMID: 39333851 PMCID: PMC11428336 DOI: 10.1186/s12576-024-00940-5] [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: 07/31/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
Glutinous rice (mochi rice), compared to non-glutinous rice (uruchi rice), exhibits a wide range of glycemic index (GI) values, from low to high. However, the underlying mechanisms behind the variation in GI values remain poorly understood. In this study, we aimed to identify rice cultivars with a low postprandial glycemic response and investigate the mechanisms, focusing on insulin and incretin hormones. We examined seven glutinous rice cultivars and three non-glutinous rice cultivars. We discovered that Anekomochi, a glutinous rice cultivar, has the lowest postprandial glycemic response. Anekomochi significantly enhanced glucagon-like peptide-1 (GLP-1) secretion while suppressing insulin secretion. These effects were completely blunted by inhibiting GLP-1 receptor signaling and denervating the common hepatic branch of vagal afferent nerves that are crucial for sensing intestinal GLP-1. Our findings demonstrate that Anekomochi markedly enhances insulin action via GLP-1 release and vagal afferent neural pathways, thereby leading to a lower postprandial glycemic response.
Collapse
Affiliation(s)
- Kento Ohbayashi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Yudai Sugiyama
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Taichi Nohmi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Kazusa Nishimura
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, 1-1-1 Tsushimanaka, Kita-Ku, Okayama, 700-0082, Japan
- Graduate School of Agriculture, Kyoto University, 4-2-1, Shiroyamadai, Kizugawa, Kyoto, 619-0218, Japan
| | - Tetsuya Nakazaki
- Graduate School of Agriculture, Kyoto University, 4-2-1, Shiroyamadai, Kizugawa, Kyoto, 619-0218, Japan
- Office of Institutional Advancement and Communications, Kyoto University, Yoshida-Honmachi, Sakyo-Ku, Kyoto, 606-8501, Japan
| | - Yo-Ichiro Sato
- Research Center for Japanese Food Culture, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
- Museum of Natural and Environmental History, Shizuoka, 5762 Oya, Suruga-Ku, Shizuoka, 422-8017, Japan
| | - Takehiro Masumura
- Laboratory of Genetic Engineering, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-Cho, Shimogamo, Sakyo-Ku, Kyoto, 606-8522, Japan.
| |
Collapse
|
8
|
Nishida K, Ueno S, Seino Y, Hidaka S, Murao N, Asano Y, Fujisawa H, Shibata M, Takayanagi T, Ohbayashi K, Iwasaki Y, Iizuka K, Okuda S, Tanaka M, Fujii T, Tochio T, Yabe D, Yamada Y, Sugimura Y, Hirooka Y, Hayashi Y, Suzuki A. Impaired Fat Absorption from Intestinal Tract in High-Fat Diet Fed Male Mice Deficient in Proglucagon-Derived Peptides. Nutrients 2024; 16:2270. [PMID: 39064713 PMCID: PMC11280123 DOI: 10.3390/nu16142270] [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: 06/19/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Proglucagon-derived peptides (PDGPs) including glucagon (Gcg), GLP-1, and GLP-2 regulate lipid metabolism in the liver, adipocytes, and intestine. However, the mechanism by which PGDPs participate in alterations in lipid metabolism induced by high-fat diet (HFD) feeding has not been elucidated. (2) Methods: Mice deficient in PGDP (GCGKO) and control mice were fed HFD for 7 days and analyzed, and differences in lipid metabolism in the liver, adipose tissue, and duodenum were investigated. (3) Results: GCGKO mice under HFD showed lower expression levels of the genes involved in free fatty acid (FFA) oxidation such as Hsl, Atgl, Cpt1a, Acox1 (p < 0.05), and Pparα (p = 0.05) mRNA in the liver than in control mice, and both FFA and triglycerides content in liver and adipose tissue weight were lower in the GCGKO mice. On the other hand, phosphorylation of hormone-sensitive lipase (HSL) in white adipose tissue did not differ between the two groups. GCGKO mice under HFD exhibited lower expression levels of Pparα and Cd36 mRNA in the duodenum as well as increased fecal cholesterol contents compared to HFD-controls. (4) Conclusions: GCGKO mice fed HFD exhibit a lesser increase in hepatic FFA and triglyceride contents and adipose tissue weight, despite reduced β-oxidation in the liver, than in control mice. Thus, the absence of PGDP prevents dietary-induced fatty liver development due to decreased lipid uptake in the intestinal tract.
Collapse
Affiliation(s)
- Koki Nishida
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Shinji Ueno
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Yusuke Seino
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
| | - Shihomi Hidaka
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Naoya Murao
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
| | - Yuki Asano
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Haruki Fujisawa
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Megumi Shibata
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Takeshi Takayanagi
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Kento Ohbayashi
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan; (K.O.); (Y.I.)
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan; (K.O.); (Y.I.)
| | - Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University, Toyoake 470-1192, Japan;
| | - Shoei Okuda
- Graduate School of Bioscience and Biotechnology, College of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (S.O.); (M.T.)
| | - Mamoru Tanaka
- Graduate School of Bioscience and Biotechnology, College of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Japan; (S.O.); (M.T.)
| | - Tadashi Fujii
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Japan; (T.F.); (T.T.); (Y.H.)
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake 470-1101, Japan
- BIOSIS Lab. Co., Ltd., Toyoake 470-1192, Japan
| | - Takumi Tochio
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Japan; (T.F.); (T.T.); (Y.H.)
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake 470-1101, Japan
- BIOSIS Lab. Co., Ltd., Toyoake 470-1192, Japan
| | - Daisuke Yabe
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
- Center for One Medicine Innovative Translational Research, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yuuichiro Yamada
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto 604-8436, Japan; (D.Y.); (Y.Y.)
| | - Yoshihisa Sugimura
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Japan; (T.F.); (T.T.); (Y.H.)
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake 470-1101, Japan
- BIOSIS Lab. Co., Ltd., Toyoake 470-1192, Japan
| | - Yoshitaka Hayashi
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan;
- Department of Endocrinology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Atsushi Suzuki
- Departments of Endocrinology, Diabetes and Metabolism, Fujita Health University School of Medicine, Toyoake 470-1192, Japan; (K.N.); (S.U.); (S.H.); (N.M.); (Y.A.); (H.F.); (M.S.); (T.T.); (Y.S.); (A.S.)
| |
Collapse
|
9
|
Pendergrast LA, Ashcroft SP, Ehrlich AM, Treebak JT, Krook A, Dollet L, Zierath JR. Metabolic plasticity and obesity-associated changes in diurnal postexercise metabolism in mice. Metabolism 2024; 155:155834. [PMID: 38479569 DOI: 10.1016/j.metabol.2024.155834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Circadian disruption is widespread and increases the risk of obesity. Timing of therapeutic interventions may promote coherent and efficient gating of metabolic processes and restore energy homeostasis. AIM To characterize the diurnal postexercise metabolic state in mice and to identify the influence of diet-induced obesity on identified outcomes. METHODS C57BL6/NTac male mice (6 wks of age) were fed a standard chow or high-fat diet for 5 weeks. At week 5, mice were subjected to a 60-min (16 m/min, 5 % incline) running bout (or sham) during the early rest (day) or early active (night) phase. Tissue and serum samples were collected immediately post-exercise (n = 6/group). In vivo glucose oxidation was measured after oral administration of 13C-glucose via 13CO2 exhalation analysis in metabolic cages. Basal and isoproterenol-stimulated adipose tissue lipolysis was assessed ex vivo for 1 h following exercise. RESULTS Lean mice displayed exercise-timing-specific plasticity in metabolic outcomes, including phase-specificity in systemic glucose metabolism and adipose-tissue-autonomous lipolytic activity depending on time of day. Conversely, obesity impaired temporal postexercise differences in whole-body glucose oxidation, as well as the phase- and exercise-mediated induction of lipolysis in isolated adipose tissue. This obesity-induced alteration in diurnal metabolism, as well as the indistinct response to exercise, was observed concomitant with disruption of core clock gene expression in peripheral tissues. CONCLUSIONS Overall, high-fat fed obese mice exhibit metabolic inflexibility, which is also evident in the diurnal exercise response. Our study provides physiological insight into exercise timing-dependent aspects in the dynamic regulation of metabolism and the influence of obesity on this biology.
Collapse
Affiliation(s)
- Logan A Pendergrast
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Stephen P Ashcroft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Krook
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
| | - Lucile Dollet
- Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden; Department of Physiology and Pharmacology, Section for Integrative Physiology, Karolinska Institutet, Stockholm, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
10
|
Fehsel K, Bouvier ML. Sex-Specific Effects of Long-Term Antipsychotic Drug Treatment on Adipocyte Tissue and the Crosstalk to Liver and Brain in Rats. Int J Mol Sci 2024; 25:2188. [PMID: 38396865 PMCID: PMC10889281 DOI: 10.3390/ijms25042188] [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: 01/17/2024] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Antipsychotic drug (APD) medication can lead to metabolic dysfunctions and weight gain, which together increase morbidity and mortality. Metabolically active visceral adipose tissue (VAT) in particular plays a crucial role in the etiopathology of these metabolic dysregulations. Here, we studied the effect of 12 weeks of drug medication by daily oral feeding of clozapine and haloperidol on the perirenal fat tissue as part of VAT of male and female Sprague Dawley rats in the context of complex former investigations on brain, liver, and blood. Adipocyte area values were determined, as well as triglycerides, non-esterified fatty acids (NEFAs), glucose, glycogen, lactate, malondialdehyde equivalents, ferric iron and protein levels of Perilipin-A, hormone-sensitive-lipase (HSL), hepcidin, glucose transporter-4 (Glut-4) and insulin receptor-ß (IR-ß). We found increased adipocyte mass in males, with slightly higher adipocyte area values in both males and females under clozapine treatment. Triglycerides, NEFAs, glucose and oxidative stress in the medicated groups were unchanged or slightly decreased. In contrast to controls and haloperidol-medicated rats, perirenal adipocyte mass and serum leptin levels were not correlated under clozapine. Protein expressions of perilipin-A, Glut-4 and HSL were decreased under clozapine treatment. IR-ß expression changed sex-specifically in the clozapine-medicated groups associated with higher hepcidin levels in the perirenal adipose tissue of clozapine-treated females. Taken together, clozapine and haloperidol had a smaller effect than expected on perirenal adipose tissue. The perirenal adipose tissue shows only weak changes in lipid and glucose metabolism. The main changes can be seen in the proteins examined, and probably in their effect on liver metabolism.
Collapse
Affiliation(s)
- Karin Fehsel
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine-University, Bergische Landstraße 2, 40629 Düsseldorf, Germany;
| | | |
Collapse
|
11
|
Ma T, Bennett T, Lee CD, Wicklow M. The diurnal pattern of moderate-to-vigorous physical activity and obesity: a cross-sectional analysis. Obesity (Silver Spring) 2023; 31:2638-2647. [PMID: 37661938 DOI: 10.1002/oby.23851] [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: 02/28/2023] [Revised: 05/25/2023] [Accepted: 06/01/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE Moderate-to-vigorous physical activity (MVPA) is obesity-protective. However, the optimal time of the day to engage in MVPA for weight management is controversial. This study is designed to investigate the influence of the diurnal pattern of MVPA on the association between MVPA and obesity. METHODS A total of 5285 participants in the 2003 to 2006 National Health and Nutrition Examination Survey (NHANES) were cross-sectionally analyzed. The diurnal pattern of objectively measured MVPA was classified into three clusters by K-means clustering analysis: morning (n = 642); midday (n = 2456); and evening (n = 2187). The associations of MVPA level and the diurnal pattern with obesity were tested. RESULTS A strong linear association between MVPA and obesity was found in the morning group, whereas a weaker curvilinear association between MVPA and obesity was observed in the midday and evening groups, respectively. Among those who met the physical activity guidelines, the adjusted means for BMI were 25.9 (95% CI: 25.2-26.6), 27.6 (95% CI: 27.1-28.1), and 27.2 (95% CI: 26.8-27.7) kg/m2 in the morning, midday, and evening groups, respectively, and for waist circumference were 91.5 (95% CI: 89.4-93.6), 95.8 (95% CI: 94.7-96.9), and 95.0 (95% CI: 93.9-96.1) cm, respectively. CONCLUSIONS The diurnal pattern of MVPA influences the association between MVPA and obesity. The promising role of morning MVPA for weight management warrants further investigation.
Collapse
Affiliation(s)
- Tongyu Ma
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
- Health Sciences Department, Franklin Pierce University, Rindge, New Hampshire, USA
| | - Thomas Bennett
- Health Sciences Department, Franklin Pierce University, Rindge, New Hampshire, USA
| | - Chong-Do Lee
- College of Health Solutions, Arizona State University, Tempe, Arizona, USA
| | - Mairead Wicklow
- Health Sciences Department, Franklin Pierce University, Rindge, New Hampshire, USA
| |
Collapse
|
12
|
Matsukawa T, Yagi T, Uchida T, Sakai M, Mitsushima M, Naganuma T, Yano H, Inaba Y, Inoue H, Yanagida K, Uematsu M, Nakao K, Nakao H, Aiba A, Nagashima Y, Kubota T, Kubota N, Izumida Y, Yahagi N, Unoki-Kubota H, Kaburagi Y, Asahara SI, Kido Y, Shindou H, Itoh M, Ogawa Y, Minami S, Terauchi Y, Tobe K, Ueki K, Kasuga M, Matsumoto M. Hepatic FASN deficiency differentially affects nonalcoholic fatty liver disease and diabetes in mouse obesity models. JCI Insight 2023; 8:e161282. [PMID: 37681411 PMCID: PMC10544238 DOI: 10.1172/jci.insight.161282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes are interacting comorbidities of obesity, and increased hepatic de novo lipogenesis (DNL), driven by hyperinsulinemia and carbohydrate overload, contributes to their pathogenesis. Fatty acid synthase (FASN), a key enzyme of hepatic DNL, is upregulated in association with insulin resistance. However, the therapeutic potential of targeting FASN in hepatocytes for obesity-associated metabolic diseases is unknown. Here, we show that hepatic FASN deficiency differentially affects NAFLD and diabetes depending on the etiology of obesity. Hepatocyte-specific ablation of FASN ameliorated NAFLD and diabetes in melanocortin 4 receptor-deficient mice but not in mice with diet-induced obesity. In leptin-deficient mice, FASN ablation alleviated hepatic steatosis and improved glucose tolerance but exacerbated fed hyperglycemia and liver dysfunction. The beneficial effects of hepatic FASN deficiency on NAFLD and glucose metabolism were associated with suppression of DNL and attenuation of gluconeogenesis and fatty acid oxidation, respectively. The exacerbation of fed hyperglycemia by FASN ablation in leptin-deficient mice appeared attributable to impairment of hepatic glucose uptake triggered by glycogen accumulation and citrate-mediated inhibition of glycolysis. Further investigation of the therapeutic potential of hepatic FASN inhibition for NAFLD and diabetes in humans should thus consider the etiology of obesity.
Collapse
Affiliation(s)
- Toshiya Matsukawa
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Takashi Yagi
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Tohru Uchida
- Department of Nutrition Management, Faculty of Health Science, Hyogo University, Kakogawa, Hyogo, Japan
| | - Mashito Sakai
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Masaru Mitsushima
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Takao Naganuma
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Hiroyuki Yano
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Yuka Inaba
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, and
- Department of Physiology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroshi Inoue
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, and
- Department of Physiology and Metabolism, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | | | - Kazuki Nakao
- Institute of Experimental Animal Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Harumi Nakao
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, School of Medicine, Tokyo Women’s Medical University, Tokyo, Japan
| | - Tetsuya Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
- Department of Clinical Nutrition, National Institutes of Biomedical Innovation, Health, and Nutrition (NIBIOHN), Tokyo, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Nutrition Therapy, The University of Tokyo, Tokyo, Japan
| | - Yoshihiko Izumida
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoya Yahagi
- Nutrigenomics Research Group, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Unoki-Kubota
- Department of Diabetic Complications, Diabetes Research Center, Research Institute, NCGM, Tokyo, Japan
| | - Yasushi Kaburagi
- Department of Diabetic Complications, Diabetes Research Center, Research Institute, NCGM, Tokyo, Japan
| | - Shun-ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, and
- Division of Medical Chemistry, Department of Metabolism and Disease, Kobe University Graduate School of Health Sciences, Kobe, Hyogo, Japan
| | - Hideo Shindou
- Department of Lipid Life Science, NCGM, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Michiko Itoh
- Department of Metabolic Syndrome and Nutritional Science, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shiro Minami
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama-shi, Toyama, Japan
| | - Kohjiro Ueki
- Department of Molecular Diabetic Medicine, Diabetes Research Center, Research Institute, NCGM, Tokyo, Japan
| | - Masato Kasuga
- The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
| | - Michihiro Matsumoto
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
- Course of Advanced and Specialized Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Veelen A, Andriessen C, Op den Kamp Y, Erazo-Tapia E, de Ligt M, Mevenkamp J, Jörgensen JA, Moonen-Kornips E, Schaart G, Esterline R, Havekes B, Oscarsson J, Schrauwen-Hinderling VB, Phielix E, Schrauwen P. Effects of the sodium-glucose cotransporter 2 inhibitor dapagliflozin on substrate metabolism in prediabetic insulin resistant individuals: A randomized, double-blind crossover trial. Metabolism 2023; 140:155396. [PMID: 36592688 DOI: 10.1016/j.metabol.2022.155396] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS Sodium-glucose cotransporter 2 inhibitor (SGLT2i) treatment in type 2 diabetes mellitus patients results in glucosuria, causing an energy loss, and triggers beneficial metabolic adaptations. It is so far unknown if SGLT2i exerts beneficial metabolic effects in prediabetic insulin resistant individuals, yet this is of interest since SGLT2is also reduce the risk for progression of heart failure and chronic kidney disease in patients without diabetes. METHODS Fourteen prediabetic insulin resistant individuals (BMI: 30.3 ± 2.1 kg/m2; age: 66.3 ± 6.2 years) underwent 2-weeks of treatment with dapagliflozin (10 mg/day) or placebo in a randomized, placebo-controlled, cross-over design. Outcome parameters include 24-hour and nocturnal substrate oxidation, and twenty-four-hour blood substrate and insulin levels. Hepatic glycogen and lipid content/composition were measured by MRS. Muscle biopsies were taken to measure mitochondrial oxidative capacity and glycogen and lipid content. RESULTS Dapagliflozin treatment resulted in a urinary glucose excretion of 36 g/24-h, leading to a negative energy and fat balance. Dapagliflozin treatment resulted in a higher 24-hour and nocturnal fat oxidation (p = 0.043 and p = 0.039, respectively), and a lower 24-hour carbohydrate oxidation (p = 0.048). Twenty-four-hour plasma glucose levels were lower (AUC; p = 0.016), while 24-hour free fatty acids and nocturnal β-hydroxybutyrate levels were higher (AUC; p = 0.002 and p = 0.012, respectively) after dapagliflozin compared to placebo. Maximal mitochondrial oxidative capacity was higher after dapagliflozin treatment (dapagliflozin: 87.6 ± 5.4, placebo: 78.1 ± 5.5 pmol/mg/s, p = 0.007). Hepatic glycogen and lipid content were not significantly changed by dapagliflozin compared to placebo. However, muscle glycogen levels were numerically higher in the afternoon in individuals on placebo (morning: 332.9 ± 27.9, afternoon: 368.8 ± 13.1 nmol/mg), while numerically lower in the afternoon on dapagliflozin treatment (morning: 371.7 ± 22.8, afternoon: 340.5 ± 24.3 nmol/mg). CONCLUSIONS/INTERPRETATION Dapagliflozin treatment of prediabetic insulin resistant individuals for 14 days resulted in significant metabolic adaptations in whole-body and skeletal muscle substrate metabolism despite being weight neutral. Dapagliflozin improved fat oxidation and ex vivo skeletal muscle mitochondrial oxidative capacity, mimicking the effects of calorie restriction. TRIAL REGISTRATION ClinicalTrials.gov NCT03721874.
Collapse
Affiliation(s)
- Anna Veelen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Charlotte Andriessen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Yvo Op den Kamp
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Edmundo Erazo-Tapia
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marlies de Ligt
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Julian Mevenkamp
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Johanna A Jörgensen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Esther Moonen-Kornips
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Gert Schaart
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Russell Esterline
- BioPharmaceuticals R&D, Late-Stage Development, Cardiovascular, Renal and Metabolism, AstraZeneca, Gaithersburg, MD, USA
| | - Bas Havekes
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Internal Medicine, Division of Endocrinology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jan Oscarsson
- BioPharmaceuticals R&D, Late-Stage Development, Cardiovascular, Renal and Metabolism, AstraZeneca, Gothenburg, Sweden
| | - Vera B Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|
14
|
Iwayama K, Seol J, Tokuyama K. Exercise Timing Matters for Glycogen Metabolism and Accumulated Fat Oxidation over 24 h. Nutrients 2023; 15:1109. [PMID: 36904109 PMCID: PMC10005671 DOI: 10.3390/nu15051109] [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: 02/01/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Due to increasingly diverse lifestyles, exercise timings vary between individuals: before breakfast, in the afternoon, or in the evening. The endocrine and autonomic nervous systems, which are associated with metabolic responses to exercise, show diurnal variations. Moreover, physiological responses to exercise differ depending on the timing of the exercise. The postabsorptive state is associated with greater fat oxidation during exercise compared to the postprandial state. The increase in energy expenditure persists during the post-exercise period, known as "Excess Post-exercise Oxygen Consumption". A 24 h evaluation of accumulated energy expenditure and substrate oxidation is required to discuss the role of exercise in weight control. Using a whole-room indirect calorimeter, researchers revealed that exercise performed during the postabsorptive state, but not during the postprandial state, increased accumulated fat oxidation over 24 h. The time course of the carbohydrate pool, as estimated by indirect calorimetry, suggests that glycogen depletion after postabsorptive exercise underlies an increase in accumulated fat oxidation over 24 h. Subsequent studies using 13C magnetic resonance spectroscopy confirmed that the variations in muscle and liver glycogen caused by postabsorptive or postprandial exercise were consistent with indirect calorimetry data. These findings suggest that postabsorptive exercise alone effectively increases 24 h fat oxidation.
Collapse
Affiliation(s)
- Kaito Iwayama
- Faculty of Budo and Sport Studies, Tenri University, Nara 632-0071, Japan
| | - Jaehoon Seol
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan
| | - Kumpei Tokuyama
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
15
|
Abstract
The circadian clock is a cell-autonomous transcription-translation feedback mechanism that anticipates and adapts physiology and behavior to different phases of the day. A variety of factors including hormones, temperature, food-intake, and exercise can act on tissue-specific peripheral clocks to alter the expression of genes that influence metabolism, all in a time-of-day dependent manner. The aim of this study was to elucidate the effects of exercise timing on adipose tissue metabolism. We performed RNA sequencing on inguinal adipose tissue of mice immediately following maximal exercise or sham treatment at the early rest or early active phase. Only during the early active phase did exercise elicit an immediate increase in serum nonesterified fatty acids. Furthermore, early active phase exercise increased expression of markers of thermogenesis and mitochondrial proliferation in inguinal adipose tissue. In vitro, synchronized 3T3-L1 adipocytes showed a timing-dependent difference in Adrb2 expression, as well as a greater lipolytic activity. Thus, the response of adipose tissue to exercise is time-of-day sensitive and may be partly driven by the circadian clock. To determine the influence of feeding state on the time-of-day response to exercise, we replicated the experiment in 10-h-fasted early rest phase mice to mimic the early active phase metabolic status. A 10-h fast led to a similar lipolytic response as observed after active phase exercise but did not replicate the transcriptomic response, suggesting that the observed changes in gene expression are not driven by feeding status. In conclusion, acute exercise elicits timing-specific effects on adipose tissue to maintain metabolic homeostasis.
Collapse
|
16
|
Dong M, Chen H, Wen S, Yuan Y, Yang L, Li Y, Yuan X, Xu D, Zhou L. The Neuronal and Non-Neuronal Pathways of Sodium-Glucose Cotransporter-2 Inhibitor on Body Weight-Loss and Insulin Resistance. Diabetes Metab Syndr Obes 2023; 16:425-435. [PMID: 36820270 PMCID: PMC9938665 DOI: 10.2147/dmso.s399367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
With the emergence of sodium-glucose cotransporter 2 inhibitors (SGLT2i), the treatment of type 2 diabetes mellitus (T2DM) has achieved a new milestone, of which the insulin-independent mechanism could produce weight loss, improve insulin resistance (IR) and exert other protective effects. Besides the well-acknowledged biochemical processes, the dysregulated balance between sympathetic and parasympathetic activity may play a significant role in IR and obesity. Weight loss caused by SGLT-2i could be achieved via activating the liver-brain-adipose neural axis in adipocytes. We previously demonstrated that SGLT-2 are widely expressed in central nervous system (CNS) tissues, and SGLT-2i could inhibit central areas associated with autonomic control through unidentified pathways, indicating that the role of the central sympathetic inhibition of SGLT-2i on blood pressure and weight loss. However, the exact pathway of SGLT2i related to these effects and to what extent it depends on the neural system are not fully understood. The evidence of how SGLT-2i interacts with the nervous system is worth exploring. Therefore, in this review, we will illustrate the potential neurological processes by which SGLT2i improves IR in skeletal muscle, liver, adipose tissue, and other insulin-target organs via the CNS and sympathetic nervous system/parasympathetic nervous system (SNS/PNS).
Collapse
Affiliation(s)
- Meiyuan Dong
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Huiling Chen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yue Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Liling Yang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Yanyan Li
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Ligang Zhou
- Graduate School of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou, Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China, Tel +8613611927616, Email
| |
Collapse
|
17
|
Paul D, Nedelcu AM. The underexplored links between cancer and the internal body climate: Implications for cancer prevention and treatment. Front Oncol 2022; 12:1040034. [PMID: 36620608 PMCID: PMC9815514 DOI: 10.3389/fonc.2022.1040034] [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: 09/08/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
In order to effectively manage and cure cancer we should move beyond the general view of cancer as a random process of genetic alterations leading to uncontrolled cell proliferation or simply a predictable evolutionary process involving selection for traits that increase cell fitness. In our view, cancer is a systemic disease that involves multiple interactions not only among cells within tumors or between tumors and surrounding tissues but also with the entire organism and its internal "milieu". We define the internal body climate as an emergent property resulting from spatial and temporal interactions among internal components themselves and with the external environment. The body climate itself can either prevent, promote or support cancer initiation and progression (top-down effect; i.e., body climate-induced effects on cancer), as well as be perturbed by cancer (bottom-up effect; i.e., cancer-induced body climate changes) to further favor cancer progression and spread. This positive feedback loop can move the system towards a "cancerized" organism and ultimately results in its demise. In our view, cancer not only affects the entire system; it is a reflection of an imbalance of the entire system. This model provides an integrated framework to study all aspects of cancer as a systemic disease, and also highlights unexplored links that can be altered to both prevent body climate changes that favor cancer initiation, progression and dissemination as well as manipulate or restore the body internal climate to hinder the success of cancer inception, progression and metastasis or improve therapy outcomes. To do so, we need to (i) identify cancer-relevant factors that affect specific climate components, (ii) develop 'body climate biomarkers', (iii) define 'body climate scores', and (iv) develop strategies to prevent climate changes, stop or slow the changes, or even revert the changes (climate restoration).
Collapse
Affiliation(s)
- Doru Paul
- Weill Cornell Medicine, New York, NY, United States
| | - Aurora M. Nedelcu
- Biology Department, University of New Brunswick, Fredericton, NB, Canada
| |
Collapse
|
18
|
Kim HK, Radak Z, Takahashi M, Inami T, Shibata S. Chrono-exercise: Time-of-day-dependent physiological responses to exercise. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 5:50-58. [PMID: 36994180 PMCID: PMC10040331 DOI: 10.1016/j.smhs.2022.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/10/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Exercise is an effective strategy to prevent and improve obesity and related metabolic diseases. Exercise increases the metabolic demand in the body. Although many of the metabolic health benefits of exercise depend on skeletal muscle adaptations, exercise exerts many of its metabolic effects through the liver, adipose tissue, and pancreas. Therefore, exercise is the physiological state in which inter-organ signaling is most important. By contrast, circadian rhythms in mammals are associated with the regulation of several physiological and biological functions, including body temperature, sleep-wake cycle, physical activity, hormone secretion, and metabolism, which are controlled by clock genes. Glucose and lipid tolerance reportedly exhibit diurnal variations, being lower in the evening than in the morning. Therefore, the effects of exercise on substrate metabolism at different times of the day may differ. In this review, the importance of exercise timing considerations will be outlined, incorporating a chrono-exercise perspective.
Collapse
|
19
|
Onuma S, Kinoshita S, Shimba S, Ozono K, Michigami T, Kawai M. The Lack of Bmal1, a Core Clock Gene, in the Intestine Decreases Glucose Absorption in Mice. Endocrinology 2022; 163:6651710. [PMID: 35904419 DOI: 10.1210/endocr/bqac119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Indexed: 11/19/2022]
Abstract
The circadian clock network is an evolutionarily conserved system that regulates systemic metabolism, such as glucose homeostasis. Intestinal tissue is a pivotal organ for the regulation of glucose metabolism, mainly via glucose absorption into the circulation; however, the significance of the intestinal circadian clock network for glucose metabolism remains largely unclear. We herein utilized a mouse model in which Bmal1, a core clock gene, was deleted in an intestine-specific manner (Bmal1Int-/- mice) and demonstrated a rhythmic expression of Sglt1 with its peak at zeitgeber time (ZT) 10.7 ± 2.8 in control mice, whereas this was lost in Bmal1Int-/- mice. Mechanistically, chromatin immunoprecipitation analysis revealed rhythmic binding of CLOCK to the E-box elements in the Sglt1 gene in control mice; however, this was absent in Bmal1Int-/- mice. Accordingly, SGLT1 protein levels were decreased during the dark phase in Bmal1Int-/- mice and this was associated with impaired glucose absorption, leading to a decline in hepatic glycogen levels at ZT4, which was restored by ingestion of high-sucrose water. Additionally, when mice were starved from ZT0, greater expression of the lipolysis-related gene Pnpla2 was observed in adipose tissue of Bmal1Int-/- mice, and this was not noted when glycogen storage was restored by high-sucrose water prior to fasting, suggesting that higher Pnpla2 expression in Bmal1Int-/- mice was likely caused by lower glycogen storage. These results indicate that disruption of the intestinal circadian clock system impairs glucose absorption in the intestine and affects systemic glucose homeostasis.
Collapse
Affiliation(s)
- Shinsuke Onuma
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Saori Kinoshita
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
| | - Shigeki Shimba
- Department of Health Science, School of Pharmacy, Nihon University, Funabashi, Chiba 274-8555, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
20
|
Casagrande BP, Bueno AA, Pisani LP, Estadella D. Hepatic glycogen participates in the regulation of hypothalamic pAkt/Akt ratio in high-sugar/high-fat diet-induced obesity. Metab Brain Dis 2022; 37:1423-1434. [PMID: 35316448 DOI: 10.1007/s11011-022-00944-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
The hypothalamus is a major integrating centre that controls energy homeostasis and plays a major role in hepatic glycogen (HGlyc) turnover. Not only do hypothalamic and hepatic Akt levels influence glucose homeostasis and glycogen synthesis, but exposure to high-sugar/high-fat diets (HSHF) can also lead to hypothalamic inflammation and HGlyc accumulation. HSHF withdrawal overall restores energy and glucose homeostasis, but the actual relationship between hypothalamic inflammation and HGlyc after short-term HSHF withdrawal has not yet been fully elucidated. Here we investigated the short-term effects of HSHF withdrawal preceded by a 30-day HSHF intake on the liver-hypothalamus crosstalk and glucose homeostasis. Sixty-day old male Wistar rats were fed for 30 days a control chow (n = 10) (Ct), or an HSHF diet (n = 20). On the 30th day of dietary intervention, a random HSHF subset (n = 10) had their diets switched to control chow for 48 h (Hw) whilst the remaining HSHF rats remained in the HSHF diet (n = 10) (Hd). All rats were anaesthetized and euthanized at the end of the protocol. We quantified HGlyc, Akt phosphorylation, inflammation and glucose homeostasis biomarkers. We also assessed the effect of propensity to obesity on those biomarkers, as detailed previously. Hd rats showed impaired glucose homeostasis, higher HGlyc and hypothalamic inflammation, and lower pAkt/Akt. Increased HGlyc was significantly associated with HSHF intake on pAkt/Akt lowered levels. We also found that HGlyc breakdown may have prevented a further pAkt/Akt drop after HSHF withdrawal. Propensity to obesity showed no apparent effect on hypothalamic inflammation or glucose homeostasis. Our findings suggest a comprehensive role of HGlyc as a structural and functional modulator of energy metabolism, and such roles may come into play relatively rapidly.
Collapse
Affiliation(s)
- Breno P Casagrande
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Henwick Grove, WR2 6AJ, Worcester, United Kingdom
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Debora Estadella
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil.
| |
Collapse
|
21
|
Yoshida R, Yano Y, Hoshi N, Okamoto N, Sui Y, Yamamoto A, Asaji N, Shiomi Y, Yasutomi E, Hatazawa Y, Hayashi H, Ueda Y, Kodama Y. Acid-treated high-amylose corn starch suppresses high-fat diet-induced steatosis. J Food Sci 2022; 87:2173-2184. [PMID: 35411589 DOI: 10.1111/1750-3841.16146] [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: 09/24/2021] [Revised: 02/17/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022]
Abstract
Resistant starch (RS) has been reported to improve steatosis as well as obesity. Type 4 resistant starch (RS4), a chemically modified starch, is particularly hard to digest and suggesting higher efficacy. However, because the effects of RS4 on steatosis are not yet fully understood, the effects of RS4 on steatosis were examined using a murine high-fat diet model. Seven-week-old male mice were divided into three groups and fed a normal diet, a high-fat diet (HFD), or a high-fat diet with added RS (HFD + RS). Amylofiber SH® produced from acid-treated corn starch was used as the dietary RS. At 22 weeks old, hepatic steatosis and short chain fatty acid (SCFA) content and gut microbiota in cecum stool samples were analyzed. The ratio of body weight to 7 weeks was significantly suppressed in the HFD + RS group compared to the HFD group (132.2 ± 1.4% vs. 167.2 ± 3.9%, p = 0.0076). Macroscopic and microscopic steatosis was also suppressed in the HFD + RS group. Analysis of cecum stool samples revealed elevated SCFA levels in the HFD + RS group compared with the HFD group. Metagenome analysis revealed that Bifidobacterium (17.9 ± 1.9% vs. 3.6 ± 0.7%, p = 0.0019) and Lactobacillus (14.8 ± 3.4% vs. 0.72 ± 0.23%, p = 0.0045), which degrade RS to SCFA, were more prevalent in the HFD + RS group than the HFD group. In conclusion, RS4 suppressed steatosis, and increased Bifidobacterium and Lactobacillus, and SCFAs. RS4 may prevent steatosis by modulating the intestinal environment.
Collapse
Affiliation(s)
- Ryutaro Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Yoshihiko Yano
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Namiko Hoshi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Norihiro Okamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Yunlong Sui
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Atsushi Yamamoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Naoki Asaji
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Yuuki Shiomi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Eiichiro Yasutomi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Yuri Hatazawa
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Hiroki Hayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Yoshihide Ueda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine
| |
Collapse
|
22
|
Hu S, Luo L, Zeng L. Tea combats circadian rhythm disorder syndrome via the gut-liver-brain axis: potential mechanisms speculated. Crit Rev Food Sci Nutr 2022; 63:7126-7147. [PMID: 35187990 DOI: 10.1080/10408398.2022.2040945] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Circadian rhythm is an intrinsic mechanism developed by organisms to adapt to external environmental signals. Nowadays, owing to the job and after-work entertainment, staying up late - Circadian rhythm disorders (CRD) are common. CRD is linked to the development of fatty liver, type 2 diabetes, and chronic gastroenteritis, which affecting the body's metabolic and inflammatory responses via multi-organ crosstalk (gut-liver-brain axis, etc.). However, studies on the mechanisms of multi-organ interactions by CRD are still weak. Current studies on therapeutic agents for CRD remain inadequate, and phytochemicals have been shown to alleviate CRD-induced syndromes that may be used for CRD-therapy in the future. Tea, a popular phytochemical-rich beverage, reduces glucolipid metabolism and inflammation. But it is immature and unclear in the mechanisms of alleviation of CRD-mediated syndrome. Here, we have analyzed the threat of CRD to hosts and their offspring' health from the perspective of the "gut-liver-brain" axis. The potential mechanisms of tea in alleviating CRD were further explored. It might be by interfering with bile acid metabolism, tryptophan metabolism, and G protein-coupled receptors, with FXR, AHR, and GPCR as potential targets. We hope to provide new perspectives on the role of tea in the prevention and mitigation of CRD.HighlightsThe review highlights the health challenges of CRD via the gut-liver-brain axis.CRD research should focus on the health effects on healthy models and its offspring.Tea may prevent CRD by regulating bile acid, tryptophan, and GPCR.Potential targets for tea prevention and mitigation of CRD include FXR, AHR and GPCR.A comprehensive assessment mechanism for tea in improving CRD should be established.
Collapse
Affiliation(s)
- Shanshan Hu
- College of Food Science, Southwest University, Beibei, Chongqing, People's Republic of China
| | - Liyong Luo
- College of Food Science, Southwest University, Beibei, Chongqing, People's Republic of China
| | - Liang Zeng
- College of Food Science, Southwest University, Beibei, Chongqing, People's Republic of China
| |
Collapse
|
23
|
Hänschke L, Heier C, Maya Palacios SJ, Özek HE, Thiele C, Bauer R, Kühnlein RP, Bülow MH. Drosophila Lipase 3 Mediates the Metabolic Response to Starvation and Aging. FRONTIERS IN AGING 2022; 3:800153. [PMID: 35821816 PMCID: PMC9261307 DOI: 10.3389/fragi.2022.800153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/13/2022] [Indexed: 05/23/2023]
Abstract
The human LIPA gene encodes for the enzyme lysosomal acid lipase, which hydrolyzes cholesteryl ester and triacylglycerol. Lysosomal acid lipase deficiency results in Wolman disease and cholesteryl ester storage disease. The Drosophila genome encodes for two LIPA orthologs, Magro and Lipase 3. Magro is a gut lipase that hydrolyzes triacylglycerides, while Lipase 3 lacks characterization based on mutant phenotypes. We found previously that Lipase 3 transcription is highly induced in mutants with defects in peroxisome biogenesis, but the conditions that allow a similar induction in wildtypic flies are not known. Here we show that Lipase 3 is drastically upregulated in starved larvae and starved female flies, as well as in aged male flies. We generated a lipase 3 mutant that shows sex-specific starvation resistance and a trend to lifespan extension. Using lipidomics, we demonstrate that Lipase 3 mutants accumulate phosphatidylinositol, but neither triacylglycerol nor diacylglycerol. Our study suggests that, in contrast to its mammalian homolog LIPA, Lipase 3 is a putative phospholipase that is upregulated under extreme conditions like prolonged nutrient deprivation and aging.
Collapse
Affiliation(s)
- Lea Hänschke
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Christoph Heier
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed- Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | | | - Huseyin Erdem Özek
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Christoph Thiele
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Reinhard Bauer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Ronald P. Kühnlein
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed- Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Margret H. Bülow
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
24
|
Matsubara Y, Kiyohara H, Teratani T, Mikami Y, Kanai T. Organ and brain crosstalk: The liver-brain axis in gastrointestinal, liver, and pancreatic diseases. Neuropharmacology 2021; 205:108915. [PMID: 34919906 DOI: 10.1016/j.neuropharm.2021.108915] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
The liver is the largest organ in the human body and is responsible for the metabolism and storage of the three principal nutrients: carbohydrates, fats, and proteins. In addition, the liver contributes to the breakdown and excretion of alcohol, medicinal agents, and toxic substances and the production and secretion of bile. In addition to its role as a metabolic centre, the liver has recently attracted attention for its function in the liver-brain axis, which interacts closely with the central nervous system via the autonomic nervous system, including the vagus nerve. The liver-brain axis influences the control of eating behaviour in the central nervous system through stimuli from the liver. Conversely, neural signals from the central nervous system influence glucose, lipid, and protein metabolism in the liver. The liver also receives a constant influx of nutrients and hormones from the intestinal tract and compounds of bacterial origin via the portal system. As a result, the intestinal tract and liver are involved in various immunological interactions. A good example is the co-occurrence of primary sclerosing cholangitis and ulcerative colitis. These heterogeneous roles of the liver-brain axis are mediated via the vagus nerve in an asymmetrical manner. In this review, we provide an overview of these interactions, mainly with the liver but also with the brain and gut.
Collapse
Affiliation(s)
- Yuta Matsubara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroki Kiyohara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan.
| |
Collapse
|
25
|
Iwayama K, Tanabe Y, Tanji F, Ohnishi T, Takahashi H. Diurnal variations in muscle and liver glycogen differ depending on the timing of exercise. J Physiol Sci 2021; 71:35. [PMID: 34802419 PMCID: PMC10717652 DOI: 10.1186/s12576-021-00821-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022]
Abstract
It has been suggested that glycogen functions not only in carbohydrate energy storage, but also as molecular sensors capable of activating lipolysis. This study aimed to compare the variation in liver and muscle glycogen during the day due to different timing of exercise. Nine healthy young men participated in two trials in which they performed a single bout of exercise at 70% of their individual maximal oxygen uptake for 60 min in the post-absorptive (morning) or post-prandial (afternoon) state. Liver and muscles glycogen levels were measured using carbon magnetic resonance spectroscopy (13C MRS). Diurnal variations in liver and muscle glycogen compared to baseline levels were significantly different depending on the timing of exercise. The effect of the timing of exercise on glycogen fluctuation is known to be related to a variety of metabolic signals, and the results of this study will be useful for future research on energy metabolism.
Collapse
Affiliation(s)
- Kaito Iwayama
- Faculty of Budo and Sport Studies, Tenri University, 80 Tainoshocho, Tenri, Nara, 632-0071, Japan.
| | - Yoko Tanabe
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Fumiya Tanji
- Sport Medical Science Research Institute, Tokai University, Kanagawa, Japan
| | | | - Hideyuki Takahashi
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
26
|
Kawakubo-Yasukochi T, Yano E, Kimura S, Nishinakagawa T, Mizokami A, Hayashi Y, Hatakeyama Y, Ohe K, Yasukochi A, Nakamura S, Jimi E, Hirata M. Hepatic glycogenolysis is determined by maternal high-calorie diet via methylation of Pygl and it is modified by oteocalcin administration in mice. Mol Metab 2021; 54:101360. [PMID: 34673295 PMCID: PMC8606545 DOI: 10.1016/j.molmet.2021.101360] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/11/2021] [Accepted: 10/17/2021] [Indexed: 12/24/2022] Open
Abstract
Objective Accumulating evidence indicates that an adverse perinatal environment contributes to a higher risk of metabolic disorders in the later life of the offspring. However, the underlying molecular mechanisms remain largely unknown. Thus, we investigated the contribution of maternal high-calorie diet and osteocalcin to metabolic homeostasis in the offspring. Methods Eight-week-old C57Bl/6N female mice were mated with age-matched males and allocated randomly to three groups: a normal-diet (ND) or a high-fat, high-sucrose diet group, which was administered either saline (control) or GluOC (10 ng/g body mass) from the day of mating to that of delivery, and the dams were fed a ND after the delivery. Pups weaned at 24 days after birth were analyzed. Results A maternal high-fat, high-sucrose diet during pregnancy causes metabolic disorders in the liver of the offspring via hypermethylation of the Pygl gene, encoding glycogen phosphorylase L, which mediates hepatic glycogenolysis. The reduced expression of Pygl induced by the maternal diet causes the hepatic accumulation of glycogen and triglyceride in the offspring, which remains in adulthood. In addition, the administration of uncarboxylated osteocalcin during pregnancy upregulates Pygl expression via both direct CREBH and ATF4 and indirect epigenomic pathways, mitigating the maternal diet-induced obesity and abnormal glucose and lipid metabolism in adulthood. Conclusions We propose that maternal energy status is reflected in the hepatic glycogenolysis capacity of the offspring via epigenetic modification of Pygl and uncarboxylated osteocalcin regulates glycogenolysis. A high-calorie diet during pregnancy causes metabolic disorders in mouse offspring. These are mediated by low liver expression of Pygl encoding glycogen phosphorylase. Hypermethylation of the Pygl promoter in utero suppresses subsequent gene expression. Modification and phenotypic changes are prevented by GluOC administration during pregnancy.
Collapse
Affiliation(s)
- Tomoyo Kawakubo-Yasukochi
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Ena Yano
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Soi Kimura
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takuya Nishinakagawa
- Department of Immunological and Molecular Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Akiko Mizokami
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshikazu Hayashi
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Yuji Hatakeyama
- Division of Functional Structure, Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Kenji Ohe
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Atsushi Yasukochi
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Eijiro Jimi
- OBT Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masato Hirata
- Oral Medicine Research Center, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan.
| |
Collapse
|
27
|
Feeding Rhythm-Induced Hypothalamic Agouti-Related Protein Elevation via Glucocorticoids Leads to Insulin Resistance in Skeletal Muscle. Int J Mol Sci 2021; 22:ijms221910831. [PMID: 34639172 PMCID: PMC8509554 DOI: 10.3390/ijms221910831] [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: 08/30/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022] Open
Abstract
Circadian phase shifts in peripheral clocks induced by changes in feeding rhythm often result in insulin resistance. However, whether the hypothalamic control system for energy metabolism is involved in the feeding rhythm-related development of insulin resistance is unknown. Here, we show the physiological significance and mechanism of the involvement of the agouti-related protein (AgRP) in evening feeding-associated alterations in insulin sensitivity. Evening feeding during the active dark period increased hypothalamic AgRP expression and skeletal muscle insulin resistance in mice. Inhibiting AgRP expression by administering an antisense oligo or a glucocorticoid receptor antagonist mitigated these effects. AgRP-producing neuron-specific glucocorticoid receptor-knockout (AgRP-GR-KO) mice had normal skeletal muscle insulin sensitivity even under evening feeding schedules. Hepatic vagotomy enhanced AgRP expression in the hypothalamus even during ad-lib feeding in wild-type mice but not in AgRP-GR-KO mice. The findings of this study indicate that feeding in the late active period may affect hypothalamic AgRP expression via glucocorticoids and induce skeletal muscle insulin resistance.
Collapse
|
28
|
Adler GK, Hornik ES, Murray G, Bhandari S, Yadav Y, Heydarpour M, Basu R, Garg R, Tirosh A. Acute effects of the food preservative propionic acid on glucose metabolism in humans. BMJ Open Diabetes Res Care 2021; 9:9/1/e002336. [PMID: 34312159 PMCID: PMC8314753 DOI: 10.1136/bmjdrc-2021-002336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/30/2021] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Propionic acid (PA) is a common food preservative generally recognized as safe by the US Food and Drug Administration; however, exogenous PA has effects on glucose metabolism that are not fully understood. Our preclinical studies demonstrated exogenous PA increases glucagon, norepinephrine, and endogenous glucose production (EGP). RESEARCH DESIGN AND METHODS We performed a randomized, placebo-controlled, crossover study in 28 healthy men and women to determine the effect of PA (1500 mg calcium propionate) on these factors. Subjects had two study visits, each preceded by a 1 week, PA-free diet. During each visit, glucose, insulin, glucagon, norepinephrine, epinephrine, and EGP were assessed for 2 hours after oral administration of PA/placebo under resting conditions (protocol 1) and during either a euglycemic (~85-90 mg/dL) or hypoglycemic (~65-70 mg/dL) hyperinsulinemic clamp (protocol 2). RESULTS PA, as compared with placebo, significantly increased: (1) glucagon and norepinephrine during protocol 1; (2) glucagon, norepinephrine, and epinephrine under euglycemic conditions in protocol 2; and (3) norepinephrine, epinephrine, and EGP under hypoglycemic conditions in protocol 2. CONCLUSION Oral consumption of PA leads to inappropriate activation of the insulin counterregulatory hormonal network. This inappropriate stimulation highlights PA as a potential metabolic disruptor.
Collapse
Affiliation(s)
- Gail K Adler
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ezra S Hornik
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Gillian Murray
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Shreya Bhandari
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yogesh Yadav
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Mahyar Heydarpour
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Rita Basu
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Rajesh Garg
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Amir Tirosh
- Division of Endocrinology, Sheba Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
29
|
Nahata M, Mogami S, Sekine H, Iizuka S, Okubo N, Fujitsuka N, Takeda H. Bcl-2-dependent autophagy disruption during aging impairs amino acid utilization that is restored by hochuekkito. NPJ Aging Mech Dis 2021; 7:13. [PMID: 34210978 PMCID: PMC8249599 DOI: 10.1038/s41514-021-00065-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic undernutrition contributes to the increase in frailty observed among elderly adults, which is a pressing issue in the sector of health care for older people worldwide. Autophagy, an intracellular recycling system, is closely associated with age-related pathologies. Therefore, decreased autophagy in aging could be involved in the disruption of energy homeostasis that occurs during undernutrition; however, the physiological mechanisms underlying this process remain unknown. Here, we showed that 70% daily food restriction (FR) induced fatal hypoglycemia in 23–26-month-old (aged) mice, which exhibited significantly lower hepatic autophagy than 9-week-old (young) mice. The liver expressions of Bcl-2, an autophagy-negative regulator, and Beclin1–Bcl-2 binding, were increased in aged mice compared with young mice. The autophagy inducer Tat-Beclin1 D11, not the mTOR inhibitor rapamycin, decreased the plasma levels of the glucogenic amino acid and restored the blood glucose levels in aged FR mice. Decreased liver gluconeogenesis, body temperature, physical activity, amino acid metabolism, and hepatic mitochondrial dynamics were observed in the aged FR mice. These changes were restored by treatment with hochuekkito that is a herbal formula containing several autophagy-activating ingredients. Our results indicate that Bcl-2 upregulation in the liver during the aging process disturbs autophagy activation, which increases the vulnerability to undernutrition. The promotion of liver autophagy may offer clinical therapeutic benefits to frail elderly patients.
Collapse
Affiliation(s)
- Miwa Nahata
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Sachiko Mogami
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Hitomi Sekine
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Seiichi Iizuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Naoto Okubo
- Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Naoki Fujitsuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ibaraki, Japan
| | - Hiroshi Takeda
- Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan. .,Hokkaido University Hospital Gastroenterological Medicine, Sapporo, Hokkaido, Japan.
| |
Collapse
|
30
|
Sano A, Seki Y, Kasama K, Nabekura T, Kurokawa Y, Ubukata Y, Nakazawa N, Hara K, Sakai M, Sohda M, Shirabe K, Saeki H. Impact of the Hepatic Branch of the Vagus Nerve Transection in Laparoscopic Sleeve Gastrectomy for Patients with Obesity and Type 2 Diabetes Mellitus. Obes Surg 2021; 31:3926-3935. [PMID: 34081275 DOI: 10.1007/s11695-021-05510-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND An increase in gastroesophageal reflux disease (GERD) after laparoscopic sleeve gastrectomy (LSG) has been reported, and concomitant hiatal hernia repair (HHR) during LSG is expected to reduce the incidence of post-LSG GERD. In HHR, the hepatic branch of the vagus nerve is anatomically transected. Recent experimental animal models suggest that vagotomy may affect glycemic control and weight loss through a neuroendocrine response. OBJECTIVES To examine whether LSG with/without hepatic branch vagotomy (HV) has a clinical impact on glycemic control in patients with obesity and type 2 diabetes mellitus (T2DM). Furthermore, the impact on weight loss and post-LSG GERD were evaluated. METHODS A total of 204 Japanese patients with obesity and T2DM, who underwent LSG and completed 1-year follow-up, were retrospectively analyzed. Operative outcomes, weight loss, glycemic, and GERD-related parameters were compared between the LSG/HHR/HV group (n = 89) and the LSG group (n = 115). RESULTS There was no significant difference in the background factors in terms of anthropometric and T2DM-related parameters between the groups. The median operation times in the LSG/HHR/HV and LSG groups were 133 and 124 minutes, respectively (p = 0.236). At 1 year, the diabetes remission rate, HbA1c, fasting glucose, and C-peptide levels were all comparable between the groups. The weight loss effect was also comparable. The patients in the LSG/HHR/HV group achieved significant improvement and prevention of GERD and hiatus hernia (p < 0.001). CONCLUSION HV does not appear to have a clinical impact on glycemic control and weight loss. Concomitant HHR with LSG serves to reduce post-LSG GERD.
Collapse
Affiliation(s)
- Akihiko Sano
- Weight Loss and Metabolic Surgery Center, Yotsuya Medical Cube, 7-7 Nibancho, Chiyoda-ku, Tokyo, 102-0084, Japan.,Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Yosuke Seki
- Weight Loss and Metabolic Surgery Center, Yotsuya Medical Cube, 7-7 Nibancho, Chiyoda-ku, Tokyo, 102-0084, Japan.
| | - Kazunori Kasama
- Weight Loss and Metabolic Surgery Center, Yotsuya Medical Cube, 7-7 Nibancho, Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Taiki Nabekura
- Weight Loss and Metabolic Surgery Center, Yotsuya Medical Cube, 7-7 Nibancho, Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Yoshimochi Kurokawa
- Weight Loss and Metabolic Surgery Center, Yotsuya Medical Cube, 7-7 Nibancho, Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Yasunari Ubukata
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Nobuhiro Nakazawa
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Keigo Hara
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sakai
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ken Shirabe
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Hiroshi Saeki
- Department of General Surgical Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
31
|
Imai J, Katagiri H. Regulation of systemic metabolism by the autonomic nervous system consisting of afferent and efferent innervation. Int Immunol 2021; 34:67-79. [PMID: 33982088 DOI: 10.1093/intimm/dxab023] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Autonomic nerves, sympathetic and parasympathetic, innervate organs and modulate their functions. It has become evident that afferent and efferent signals of the autonomic nervous system play important roles in regulating systemic metabolism, thereby maintaining homeostasis at the whole-body level. Vagal afferent nerves receive signals, such as nutrients and hormones, from the peripheral organs/tissues including the gastrointestinal tract and adipose tissue then transmit these signals to the hypothalamus, thereby regulating feeding behavior. In addition to roles in controlling appetite, areas in the hypothalamus serves as regulatory centers of both sympathetic and parasympathetic efferent fibers. These efferent innervations regulate the functions of peripheral organs/tissues, such as pancreatic islets, adipose tissues and the liver, which play roles in metabolic regulation. Furthermore, recent evidence has unraveled the metabolic regulatory systems governed by autonomic nerve circuits. In these systems, afferent nerves transmit metabolic information from peripheral organs to the central nervous system (CNS) and the CNS thereby regulates the organ functions through the efferent fibers of autonomic nerves. Thus, the autonomic nervous system regulates the homeostasis of systemic metabolism, and both afferent and efferent fibers play critical roles in its regulation. In addition, several lines of evidence demonstrate the roles of the autonomic nervous system in regulating and dysregulating the immune system. This review introduces variety of neuron-mediated inter-organ cross-talk systems and organizes the current knowledge of autonomic control/coordination of systemic metabolism, focusing especially on a liver-brain-pancreatic β-cell autonomic nerve circuit, as well as highlighting the potential importance of connections with the neuronal and immune systems.
Collapse
Affiliation(s)
- Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
32
|
Abdollahi S, Kazemi A, de Souza RJ, Clark CCT, Soltani S. The effect of meal frequency on biochemical cardiometabolic factors: A systematic review and meta-analysis of randomized controlled trials. Clin Nutr 2021; 40:3170-3181. [PMID: 33485709 DOI: 10.1016/j.clnu.2020.12.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/16/2020] [Accepted: 12/25/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Although several randomized controlled trials (RCTs) have supported the beneficial effects of higher meal frequency (MF) on cardiometabolic risk factors, the putative effects of higher MF on health remain inconclusive. This study systematically reviewed the evidence from RCTs of the effect of higher compared with lower MF on the blood lipid profile, glucose homeostasis, and adipokines. METHODS PubMed, Scopus, ISI Web of Science, and the Cochrane database were searched up to October 2020 to retrieve relevant RCTs. A DerSimonian and Laird random effects model was used to pool mean differences and 95% CI for each outcome. The quality of studies and evidence was assessed through standard methods. RESULTS Twenty-one RCTs (686 participants) were included in this meta-analysis. Overall results showed a significant improvement in total cholesterol [weighted mean difference (WMD) = -6.08 mg/dl; 95% CI: -10.68, -1.48; P = 0.01; I2 = 88%], and low-density cholesterol (LDL-C) (WMD = -6.82 mg/dl; 95% CI: -10.97, -1.60; P = 0.009; I2 = 85.7%), while LDL-C to high-density cholesterol ratio (LDL-C: HDL-C) increased (WMD = 0.22; 95% CI: 0.07, 0.36; P = 0.003; I2 = 0.0%) in higher MF vs. lower MF. No significant effects were found on measures of glycemic control, apolipoproteins-A1 and B, or leptin. In subgroup analyses, higher MF significantly reduced serum triglyceride (TG), and increased HDL-C, compared with lower MF in interventions > 12 weeks, and decreased serum TC and LDL-C in healthy participants. A significant reduction in LDL-C also was observed in studies where the same foods given both arms, simply divided into different feeding occasions, and in feeding studies, following higher MF compared to lower MF. CONCLUSION Our meta-analysis found that higher, compared with lower MF may improve total cholesterol, and LDL-C. The intervention does not affect measures of glycemic control, apolipoproteins-A1 and B, or leptin. However, the GRADE ratings of low credibility of the currently available evidence highlights the need for more high-quality studies in order to reach a firm conclusion.
Collapse
Affiliation(s)
- Shima Abdollahi
- Department of Nutrition and Public Health, School of Public Health, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Asma Kazemi
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Russell J de Souza
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; Population Health Research Centre, Hamilton Health Sciences Corporation, Hamilton, ON, Canada
| | - Cain C T Clark
- Centre for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB, UK
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
33
|
John OD, Mouatt P, Panchal SK, Brown L. Rind from Purple Mangosteen ( Garcinia mangostana) Attenuates Diet-Induced Physiological and Metabolic Changes in Obese Rats. Nutrients 2021; 13:319. [PMID: 33499382 PMCID: PMC7912346 DOI: 10.3390/nu13020319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
The pulp of the purple mangosteen, Garcinia mangostana, is a popular tropical fruit but the rind containing xanthones such as α-mangostin together with procyanidins and anthocyanidins is usually discarded as waste. However, this rind has been used in South-East Asia for diarrhoea, dysentery, skin infections and wounds. As xanthones have reported anti-inflammatory and antioxidant responses, this study has determined the bioactive compounds and evaluated the effects of G. mangostana rind on physiological, metabolic, liver and cardiovascular parameters in rats with diet-induced metabolic syndrome. Rats fed a diet with increased simple sugars and saturated fats developed obesity, hypertension, increased left ventricular stiffness, dyslipidaemia and fatty liver. Administration of G. mangostana rind as 5% of the food to rats with diet-induced metabolic syndrome gave a dose of 168 mg/kg/day α-mangostin, 355 mg/kg/day procyanidins, 3.9 mg/kg/day anthocyanins and 11.8 mg/kg/day hydroxycitric acid for 8 weeks which reduced body weight and attenuated physiological and metabolic changes in rats including decreased abdominal fat deposition, decreased abdominal circumference and whole-body fat mass, improved liver structure and function and improved cardiovascular parameters such as systolic blood pressure, left ventricular stiffness and endothelial function. These responses were associated with decreased infiltration of inflammatory cells, decreased deposition of collagen in both heart and liver and decreased mean adipocyte size in retroperitoneal adipose tissues. We conclude that, in rats with diet-induced metabolic syndrome, chronic intake of G. mangostana rind decreased infiltration of inflammatory cells which decreased physiological, metabolic, liver and cardiovascular symptoms.
Collapse
Affiliation(s)
- Oliver D. John
- Functional Foods Research Group, University of Southern Queensland, Toowoomba, QLD 4350, Australia; (O.D.J.); (S.K.P.)
| | - Peter Mouatt
- Southern Cross Plant Science, Southern Cross University, Lismore, NSW 2480, Australia;
| | - Sunil K. Panchal
- Functional Foods Research Group, University of Southern Queensland, Toowoomba, QLD 4350, Australia; (O.D.J.); (S.K.P.)
| | - Lindsay Brown
- Functional Foods Research Group, University of Southern Queensland, Toowoomba, QLD 4350, Australia; (O.D.J.); (S.K.P.)
- School of Health and Wellbeing, University of Southern Queensland, Ipswich, QLD 4305, Australia
| |
Collapse
|
34
|
Evaluation of Metabolic Profiles of Patients with Anorexia Nervosa at Inpatient Admission, Short- and Long-Term Weight Regain-Descriptive and Pattern Analysis. Metabolites 2020; 11:metabo11010007. [PMID: 33374417 PMCID: PMC7823299 DOI: 10.3390/metabo11010007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/26/2022] Open
Abstract
Acute anorexia nervosa (AN) constitutes an extreme physiological state. We aimed to detect state related metabolic alterations during inpatient admission and upon short- and long-term weight regain. In addition, we tested the hypothesis that metabolite concentrations adapt to those of healthy controls (HC) after long-term weight regain. Thirty-five female adolescents with AN and 25 female HC were recruited. Based on a targeted approach 187 metabolite concentrations were detected at inpatient admission (T0), after short-term weight recovery (T1; half of target-weight) and close to target weight (T2). Pattern hunter and time course analysis were performed. The highest number of significant differences in metabolite concentrations (N = 32) were observed between HC and T1. According to the detected main pattern, metabolite concentrations at T2 became more similar to those of HC. The course of single metabolite concentrations (e.g., glutamic acid) revealed different metabolic subtypes within the study sample. Patients with AN after short-term weight regain are in a greater “metabolic imbalance” than at starvation. After long-term weight regain, patients reach a metabolite profile similar to HC. Our results might be confounded by different metabolic subtypes of patients with AN.
Collapse
|
35
|
Iwayam K, Ogawa A, Tanaka Y, Yajima K, Park I, Ando A, Ogata H, Kayaba M, Zhang S, Tanji F, Nabekura Y, Yamamoto K, Tokuyama K. Effects of exercise before breakfast on plasma free fatty acid profile and 24-h fat oxidation. Metabol Open 2020; 8:100067. [PMID: 33294835 PMCID: PMC7695873 DOI: 10.1016/j.metop.2020.100067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/25/2020] [Accepted: 11/12/2020] [Indexed: 11/29/2022] Open
Abstract
Background Free fatty acids (FFAs) are an important source of energy, and also serve as signaling molecules to regulate gene expression. Exercise performed in a post-absorptive state, in contrast to that performed in a postprandial state, increases 24-h fat oxidation under an energy-balanced condition. The primary aim of the present study was to clarify whether the effects of exercise on the concentration and composition of plasma FFAs, which may underlie distinct effects of exercise on 24-h fat oxidation, depend on the nutritional state of the individual when performing the exercise. Methods Ten healthy young men underwent 3 trials of indirect calorimetry in a metabolic chamber. The subjects performed exercise at 60% of VO2max for 60 min in either a post-absorptive or postprandial state, or remained sedentary without an exercise session (control). All trials were designed to be energy balanced over 24 h. Blood samples were collected immediately before and after exercise. Results Fat oxidation over 24 h was increased only when exercise was performed in a post-absorptive state (control, 531 ± 60; post-absorptive, 779 ± 70; postprandial, 569 ± 37 kcal/24 h). The increase in the 24-h fat oxidation was related to the magnitude of the transient carbohydrate deficit after exercise. The plasma FFA concentration after exercise was higher in the post-absorptive trial (0.38 ± 0.04) than in the control (0.13 ± 0.01) and postprandial (0.15 ± 0.02 mM) trials. The ratio of unsaturated to saturated (U/S) fatty acids after exercise was higher in the post-absorptive trial (1.76 ± 0.06) than in the control (1.56 ± 0.07) and postprandial (1.53 ± 0.08) trials. On the other hand, the plasma FFA concentration after exercise in a postprandial state did not differ significantly from that in the control trial. Conclusion Exercise performed in a post-absorptive state effectively increased the plasma FFA concentration and U/S ratio to a greater degree than exercise performed in a postprandial state, underlying the increase in the 24-h fat oxidation. The increase in the plasma FFA concentration was related to the transient carbohydrate deficit after exercise. Exercise performed in post-absorptive state increases 24-h fat oxidation. Exercise performed in a post-absorptive state increased the ratio of U/S ratio. The plasma FFA concentration was related to the transient carbohydrate deficit.
Collapse
Affiliation(s)
- Kaito Iwayam
- Faculty of Budo and Sport Studies, Tenri University, Nara, Japan
| | - Ayane Ogawa
- Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
- AIM SERVICES CO., LTD, Tokyo, Japan
| | - Yoshiaki Tanaka
- Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Katsuhiko Yajima
- Department of Nutritional Physiology, Faculty of Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Insung Park
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Ibaraki, Japan
| | - Akira Ando
- Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Hitomi Ogata
- Graduate School of Integrated Arts and Sciences, Hiroshima University, Hiroshima, Japan
| | - Momoko Kayaba
- Department of Somnology, Tokyo Medical University, Tokyo, Japan
| | - Simeng Zhang
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Ibaraki, Japan
| | - Fumiya Tanji
- Sport Medical Science Research Institute, Tokai University, Kanagawa, Japan
| | - Yoshiharu Nabekura
- Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kouhei Yamamoto
- School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka, Japan
| | - Kumpei Tokuyama
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Ibaraki, Japan
- Corresponding author.
| |
Collapse
|
36
|
Aniort J, Montaurier C, Poyet A, Meunier N, Piraud A, Aguilera D, Bouiller M, Enache I, Ali Y, Jouve C, Blot A, Farigon N, Cano N, Boirie Y, Richard R, Heng AE. Day and night changes in energy expenditure of patients on automated peritoneal dialysis. Clin Nutr 2020; 40:3454-3461. [PMID: 33288303 DOI: 10.1016/j.clnu.2020.11.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/17/2020] [Accepted: 11/22/2020] [Indexed: 10/22/2022]
Abstract
RATIONALE Automated peritoneal dialysis (APD) treatment for end-stage kidney disease affords patients a degree of autonomy in everyday life. Clinical investigations of their energy expenditure (EE) are usually based on resting EE, which could mask day and night variations in EE. The aim of this study, therefore, was to compare the components of EE in APD patients and healthy control (C) subjects. MATERIAL AND METHOD Patients treated with APD for more than 3 months were compared with C volunteers matched for age and lean body mass (LBM). Biochemical analyses were performed and body composition was determined by DEXA to adjust EE to LBM. Total EE, its different components and respiratory quotients (RQ) were measured by a gas exchange method in calorimetric chambers. Spontaneous total and activity-related EE (AEE) were also measured in free-living conditions over 4 days by a calibrated accelerometer and a heart rate monitor. RESULTS APD (n = 7) and C (n = 7) patients did not differ in age and body composition. REE did not differ between the two groups. However, prandial increase in EE adjusted for dietary energy intake was higher in APD patients (+57.5 ± 12.71 kcal/h) than in C subjects (+33.8 ± 10.5 kcal/h, p = 0.003) and nocturnal decrease in EE tended to be lower in APD patients undergoing dialysis sessions (- 4.53 ± 8.37 kcal/h) than in subjects (- 11.8 ± 7.69 kcal/h, p = 0.059). Resting RQ (0.91 ± 0.09 vs 0.81 ± 0.04, p = 0.032) and nocturnal RQ (0.91 ± 0.09 vs 0.81 ± 0.04, p = 0.032) were significantly higher in APD patients, indicating a preferential use of glucose substrate potentially absorbed across the peritoneum. AEE was lower in APD patients (595.9 ± 383.2 kcal/d) than in C subjects (1205.2 ± 370.5 kcal/d, p = 0.011). In contrast, energy intakes were not significantly different (1986 ± 465 vs 2083 ± 377 kcal/d, p = 0.677). CONCLUSION Although the two groups had identical resting EE, APD patients had a higher prandial increase in EE, a lower activity-related EE and higher resting and nocturnal RQ than healthy subjects.
Collapse
Affiliation(s)
- Julien Aniort
- CHU Clermont-Ferrand, Nephrology, Dialysis and Transplantation Department, Gabriel Montpied Hospital, Clermont-Ferrand, France; Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France.
| | - Christophe Montaurier
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France
| | - Anais Poyet
- Association Régionale d'Aide aux Urémique du Centre Ouest (ARAUCO), Bourges, France
| | - Nathalie Meunier
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France
| | - Aurélien Piraud
- CHU Clermont-Ferrand, Nephrology, Dialysis and Transplantation Department, Gabriel Montpied Hospital, Clermont-Ferrand, France
| | - Didier Aguilera
- CH Vichy, Nephrology and Dialysis Department, Jacques Lacarin Hospital, Vichy, France
| | - Marc Bouiller
- CH Puy-en-Velay, Nephrology and Dialysis Department, Emile Roux Hospital, Puy en Velay, France
| | - Ioana Enache
- AURA Auvergne (Association pour l'Utilisation du Rein artificiel), Clermont Ferrand, France
| | - Youssef Ali
- CH Montluçon, Nephrology and Dialysis Department, Montluçon hospital, Montluçon, France
| | - Christelle Jouve
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France
| | - Adeline Blot
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France
| | - Nicolas Farigon
- CHU Clermont-Ferrand, Clinical Nutrition Department, Gabriel Montpied Hospital, Clermont-Ferrand, France
| | - Noël Cano
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France; CHU Clermont-Ferrand, Clinical Nutrition Department, Gabriel Montpied Hospital, Clermont-Ferrand, France
| | - Yves Boirie
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France; CHU Clermont-Ferrand, Clinical Nutrition Department, Gabriel Montpied Hospital, Clermont-Ferrand, France
| | - Rudy Richard
- Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France; CHU Clermont-Ferrand, Sports Medicine and Functional Exploration Department, Gabriel Montpied Hospital, Clermont-Ferrand, France
| | - Anne Elisabeth Heng
- CHU Clermont-Ferrand, Nephrology, Dialysis and Transplantation Department, Gabriel Montpied Hospital, Clermont-Ferrand, France; Clermont Auvergne University, INRAE, Human Nutrition Unit, CRNH Auvergne, Clermont-Ferrand, France
| |
Collapse
|
37
|
Huang SSY, Makhlouf M, AbouMoussa EH, Ruiz Tejada Segura ML, Mathew LS, Wang K, Leung MC, Chaussabel D, Logan DW, Scialdone A, Garand M, Saraiva LR. Differential regulation of the immune system in a brain-liver-fats organ network during short-term fasting. Mol Metab 2020; 40:101038. [PMID: 32526449 PMCID: PMC7339127 DOI: 10.1016/j.molmet.2020.101038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/24/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Fasting regimens can promote health, mitigate chronic immunological disorders, and improve age-related pathophysiological parameters in animals and humans. Several ongoing clinical trials are using fasting as a potential therapy for various conditions. Fasting alters metabolism by acting as a reset for energy homeostasis, but the molecular mechanisms underlying the beneficial effects of short-term fasting (STF) are not well understood, particularly at the systems or multiorgan level. METHODS We performed RNA-sequencing in nine organs from mice fed ad libitum (0 h) or subjected to fasting five times (2-22 h). We applied a combination of multivariate analysis, differential expression analysis, gene ontology, and network analysis for an in-depth understanding of the multiorgan transcriptome. We used literature mining solutions, LitLab™ and Gene Retriever™, to identify the biological and biochemical terms significantly associated with our experimental gene set, which provided additional support and meaning to the experimentally derived gene and inferred protein data. RESULTS We cataloged the transcriptional dynamics within and between organs during STF and discovered differential temporal effects of STF among organs. Using gene ontology enrichment analysis, we identified an organ network sharing 37 common biological pathways perturbed by STF. This network incorporates the brain, liver, interscapular brown adipose tissue, and posterior-subcutaneous white adipose tissue; hence, we named it the brain-liver-fats organ network. Using Reactome pathways analysis, we identified the immune system, dominated by T cell regulation processes, as a central and prominent target of systemic modulations during STF in this organ network. The changes we identified in specific immune components point to the priming of adaptive immunity and parallel the fine-tuning of innate immune signaling. CONCLUSIONS Our study provides a comprehensive multiorgan transcriptomic profiling of mice subjected to multiple periods of STF and provides new insights into the molecular modulators involved in the systemic immunotranscriptomic changes that occur during short-term energy loss.
Collapse
Affiliation(s)
| | | | | | - Mayra L Ruiz Tejada Segura
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Marchioninistraße 25, 81377, München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| | | | - Kun Wang
- Sidra Medicine, PO Box 26999, Doha, Qatar.
| | | | | | - Darren W Logan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Marchioninistraße 25, 81377, München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| | | | - Luis R Saraiva
- Sidra Medicine, PO Box 26999, Doha, Qatar; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
38
|
Fernandes AB, Alves da Silva J, Almeida J, Cui G, Gerfen CR, Costa RM, Oliveira-Maia AJ. Postingestive Modulation of Food Seeking Depends on Vagus-Mediated Dopamine Neuron Activity. Neuron 2020; 106:778-788.e6. [PMID: 32259476 PMCID: PMC7710496 DOI: 10.1016/j.neuron.2020.03.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/07/2020] [Accepted: 03/12/2020] [Indexed: 01/07/2023]
Abstract
Postingestive nutrient sensing can induce food preferences. However, much less is known about the ability of postingestive signals to modulate food-seeking behaviors. Here we report a causal connection between postingestive sucrose sensing and vagus-mediated dopamine neuron activity in the ventral tegmental area (VTA), supporting food seeking. The activity of VTA dopamine neurons increases significantly after administration of intragastric sucrose, and deletion of the NMDA receptor in these neurons, which affects bursting and plasticity, abolishes lever pressing for postingestive sucrose delivery. Furthermore, lesions of the hepatic branch of the vagus nerve significantly impair postingestive-dependent VTA dopamine neuron activity and food seeking, whereas optogenetic stimulation of left vagus nerve neurons significantly increases VTA dopamine neuron activity. These data establish a necessary role of vagus-mediated dopamine neuron activity in postingestive-dependent food seeking, which is independent of taste signaling.
Collapse
Affiliation(s)
- Ana B. Fernandes
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,Champalimaud Clinical Centre, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,NOVA Medical School
- Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal
| | - Joaquim Alves da Silva
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,Champalimaud Clinical Centre, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,NOVA Medical School
- Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal
| | - Joana Almeida
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Durham, NC 27709, USA
| | - Charles R. Gerfen
- Laboratory of Systems Neurosciences, National Institute of Mental Health, Bethesda, MD 20814, USA
| | - Rui M. Costa
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,NOVA Medical School
- Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal,Departments of Neuroscience and Neurology, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA,Corresponding author
| | - Albino J. Oliveira-Maia
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,Champalimaud Clinical Centre, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal,NOVA Medical School
- Faculdade de Ciencias Medicas, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal,Corresponding author
| |
Collapse
|
39
|
Iwayama K, Onishi T, Maruyama K, Takahashi H. Diurnal variation in the glycogen content of the human liver using 13 C MRS. NMR IN BIOMEDICINE 2020; 33:e4289. [PMID: 32157774 DOI: 10.1002/nbm.4289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 06/10/2023]
Abstract
Glycogen in tissues functions not only as carbohydrate reserves, but also as molecular sensors capable of activating signaling pathways in response to physical activity. While glycogen in the skeletal muscles is mainly a local energy substrate, glycogen in the liver serves as a glucose reserve to maintain normal blood glucose levels in the body, even during the sleep state. The aim of this study is to compare the diurnal variation of glycogen in the muscle and liver of human subjects under normal conditions. The glycogen content was measured in the muscle and liver of 10 young, healthy, male volunteers using 13 C MRS, a non-invasive technique. The subjects remained sedentary, and glycogen concentration was measured six times daily. Experimental meals were provided to achieve individual energy balance, estimated according to the energy requirement guideline for patients from Japan. The largest variation in muscle glycogen compared with 1 h after supper (20:00 on Day 1) was 3.1 ± 8.2 mmol/L (16:00 on Day 2). In the liver, however, the glycogen content decreased by 80.6 ± 40.4 mmol/L through the overnight fasting period (07:00 on Day 2). This study demonstrated that the glycogen content in the liver was significantly lower in the morning, while the glycogen content in the calf muscles underwent minimal diurnal variation. The overnight fast is a characteristic daily condition, in which liver glycogen content is low, whereas muscle glycogen content is relatively unaffected.
Collapse
Affiliation(s)
- Kaito Iwayama
- Faculty of Budo and Sport Studies, Tenri University, Nara, Japan
| | - Takahiro Onishi
- Medical Center, Japan Institute of Sports Sciences, Tokyo, Japan
| | - Katsuya Maruyama
- MR Research & Collaboration Department, Siemens Healthcare K.K., Tokyo, Japan
| | - Hideyuki Takahashi
- Department of Sport Research, Japan Institute of Sports Sciences, Tokyo, Japan
| |
Collapse
|
40
|
Endoh M, Baba M, Endoh T, Hirayama A, Nakamura-Ishizu A, Umemoto T, Hashimoto M, Nagashima K, Soga T, Lang M, Schmidt LS, Linehan WM, Suda T. A FLCN-TFE3 Feedback Loop Prevents Excessive Glycogenesis and Phagocyte Activation by Regulating Lysosome Activity. Cell Rep 2020; 30:1823-1834.e5. [PMID: 32049013 PMCID: PMC8459211 DOI: 10.1016/j.celrep.2020.01.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 12/11/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor suppressor folliculin (FLCN) suppresses nuclear translocation of TFE3, a master transcription factor for lysosomal biogenesis, via regulation of amino-acid-sensing Rag GTPases. However, the importance of this lysosomal regulation in mammalian physiology remains unclear. Following hematopoietic-lineage-specific Flcn deletion in mice, we found expansion of vacuolated phagocytes that accumulate glycogen in their cytoplasm, phenotypes reminiscent of lysosomal storage disorder (LSD). We report that TFE3 acts in a feedback loop to transcriptionally activate FLCN expression, and FLCN loss disrupts this loop, augmenting TFE3 activity. Tfe3 deletion in Flcn knockout mice reduces the number of phagocytes and ameliorates LSD-like phenotypes. We further reveal that TFE3 stimulates glycogenesis by promoting the expression of glycogenesis genes, including Gys1 and Gyg, upon loss of Flcn. Taken together, we propose that the FLCN-TFE3 feedback loop acts as a rheostat to control lysosome activity and prevents excessive glycogenesis and LSD-like phagocyte activation.
Collapse
Affiliation(s)
- Mitsuhiro Endoh
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore; International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| | - Masaya Baba
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Tamie Endoh
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore; International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Ayako Nakamura-Ishizu
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore; International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Terumasa Umemoto
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Michihiro Hashimoto
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Kunio Nagashima
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA; Basic Science Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Singapore; International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|
41
|
Nakanishi S, Hirukawa H, Shimoda M, Tatsumi F, Kohara K, Obata A, Okauchi S, Sanada J, Fushimi Y, Mashiko A, Mune T, Kaku K, Kaneto H. Association Between Severity of Diabetic Neuropathy and Success in Weight Loss During Hospitalization Among Japanese Patients with Type 2 Diabetes: A Retrospective Observational Study. Diabetes Metab Syndr Obes 2020; 13:1669-1676. [PMID: 32523367 PMCID: PMC7237107 DOI: 10.2147/dmso.s252673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/30/2020] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION This study aimed to examine the association between severity of diabetic neuropathy and weight loss during hospitalization in overweight participants with type 2 diabetes. PATIENTS AND METHODS Participants of this study comprised 193 patients who were hospitalized for type 2 diabetes treatment. The participants were divided into two groups in the study, based on whether or not reduction of bodyweight was at least 3% during hospitalization. Using Cox models, the association between severity of neuropathy and effectiveness of weight loss under a controlled diet was analyzed. Autonomic neuropathy was assessed on patient admission by R-R interval, as measured in an electrocardiogram (CVRR), and sensory neuropathy was assessed using both 128-Hz tuning-fork vibration and Achilles tendon reflex (ATR). RESULTS The adjusted hazard ratio for weight loss of at least 3% for CVRR was 1.17 (95% confidence interval 1.07-1.28, P=0.0006) and for vibration time 1.93 (1.01-3.68, P=0.045). After dividing CVRR and vibration time into tertiles based on participant number, the adjusted hazard ratio for the high tertile of CVRR was 2.17 (1.29-3.62, P=0.003), and for the long tertile of vibration time 1.84 (1.10-3.08, P=0.02), compared with the low and short tertiles, respectively. No association was detected between ATR category and weight loss. CONCLUSION Severity of diabetic neuropathy was found to be a determinant in weight loss under a caloric restriction regimen for patients with type 2 diabetes. The results of the study suggest that the peripheral nervous system is involved in responses to medical intervention for treatment for type 2 diabetes including bodyweight management.
Collapse
Affiliation(s)
- Shuhei Nakanishi
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
- Correspondence: Shuhei Nakanishi Email
| | - Hidenori Hirukawa
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Masashi Shimoda
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Fuminori Tatsumi
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Kenji Kohara
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Atsushi Obata
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Seizo Okauchi
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Junpei Sanada
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Yoshiro Fushimi
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Akiko Mashiko
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Tomoatsu Mune
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| | - Kohei Kaku
- Department of Internal Medicine, Kawasaki Medical School, Okayama, Japan
| | - Hideaki Kaneto
- Division of Diabetes, Metabolism and Endocrinology, Kawasaki Medical School, Okayama, Japan
| |
Collapse
|
42
|
Li L, Krznar P, Erban A, Agazzi A, Martin-Levilain J, Supale S, Kopka J, Zamboni N, Maechler P. Metabolomics Identifies a Biomarker Revealing In Vivo Loss of Functional β-Cell Mass Before Diabetes Onset. Diabetes 2019; 68:2272-2286. [PMID: 31537525 DOI: 10.2337/db19-0131] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 09/10/2019] [Indexed: 11/13/2022]
Abstract
Identification of individuals with decreased functional β-cell mass is essential for the prevention of diabetes. However, in vivo detection of early asymptomatic β-cell defect remains unsuccessful. Metabolomics has emerged as a powerful tool in providing readouts of early disease states before clinical manifestation. We aimed at identifying novel plasma biomarkers for loss of functional β-cell mass in the asymptomatic prediabetes stage. Nontargeted and targeted metabolomics were applied in both lean β-Phb2-/- (β-cell-specific prohibitin-2 knockout) mice and obese db/db (leptin receptor mutant) mice, two distinct mouse models requiring neither chemical nor dietary treatments to induce spontaneous decline of functional β-cell mass promoting progressive diabetes development. Nontargeted metabolomics on β-Phb2-/- mice identified 48 and 82 significantly affected metabolites in liver and plasma, respectively. Machine learning analysis pointed to deoxyhexose sugars consistently reduced at the asymptomatic prediabetes stage, including in db/db mice, showing strong correlation with the gradual loss of β-cells. Further targeted metabolomics by gas chromatography-mass spectrometry uncovered the identity of the deoxyhexose, with 1,5-anhydroglucitol displaying the most substantial changes. In conclusion, this study identified 1,5-anhydroglucitol as associated with the loss of functional β-cell mass and uncovered metabolic similarities between liver and plasma, providing insights into the systemic effects caused by early decline in β-cells.
Collapse
Affiliation(s)
- Lingzi Li
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| | - Petra Krznar
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- PhD Program in Systems Biology, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Alexander Erban
- Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
| | - Andrea Agazzi
- Theoretical Physics Department, University of Geneva, Geneva, Switzerland
| | - Juliette Martin-Levilain
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| | - Sachin Supale
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| | - Joachim Kopka
- Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Faculty Diabetes Centre, University of Geneva Medical Centre, Geneva, Switzerland
| |
Collapse
|
43
|
Ng GYQ, Kang SW, Kim J, Alli-Shaik A, Baik SH, Jo DG, Hande MP, Sobey CG, Gunaratne J, Fann DYW, Arumugam TV. Genome-Wide Transcriptome Analysis Reveals Intermittent Fasting-Induced Metabolic Rewiring in the Liver. Dose Response 2019; 17:1559325819876780. [PMID: 31598117 PMCID: PMC6764061 DOI: 10.1177/1559325819876780] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
Scope: Intermittent fasting (IF) has been extensively reported to promote improved energy homeostasis and metabolic switching. While IF may be a plausible strategy to ameliorate the epidemiological burden of disease in many societies, our understanding of the underlying molecular mechanisms behind such effects is still lacking. The present study has sought to investigate the relationship between IF and changes in gene expression. We focused on the liver, which is highly sensitive to metabolic changes due to energy status. Mice were randomly assigned to ad libitum feeding or IF for 16 hours per day or for 24 hours on alternate days for 3 months, after which genome-wide transcriptome analysis of the liver was performed using RNA sequencing. Our findings revealed that IF caused robust transcriptomic changes in the liver that led to a complex array of metabolic changes. We also observed that the IF regimen produced distinct profiles of transcriptomic changes, highlighting the significance of temporally different periods of energy restriction. Our results suggest that IF can regulate metabolism via transcriptomic mechanisms and provide insight into how genetic interactions within the liver might lead to the numerous metabolic benefits of IF.
Collapse
Affiliation(s)
- Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sung-Wook Kang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joonki Kim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Natural Product Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Republic of Korea
| | - Asfa Alli-Shaik
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Sang-Ha Baik
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - M Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher G Sobey
- Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Jayantha Gunaratne
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.,Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
44
|
Zhang L, Yao W, Xia J, Wang T, Huang F. Glucagon-Induced Acetylation of Energy-Sensing Factors in Control of Hepatic Metabolism. Int J Mol Sci 2019; 20:ijms20081885. [PMID: 30995792 PMCID: PMC6515121 DOI: 10.3390/ijms20081885] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
The liver is the central organ of glycolipid metabolism, which regulates the metabolism of lipids and glucose to maintain energy homeostasis upon alterations of physiological conditions. Researchers formerly focused on the phosphorylation of glucagon in controlling liver metabolism. Noteworthily, emerging evidence has shown glucagon could additionally induce acetylation to control hepatic metabolism in response to different physiological states. Through inducing acetylation of complex metabolic networks, glucagon interacts extensively with various energy-sensing factors in shifting from glucose metabolism to lipid metabolism during prolonged fasting. In addition, glucagon-induced acetylation of different energy-sensing factors is involved in the advancement of nonalcoholic fatty liver disease (NAFLD) to liver cancer. Here, we summarize the latest findings on glucagon to control hepatic metabolism by inducing acetylation of energy-sensing factors. Finally, we summarize and discuss the potential impact of glucagon on the treatment of liver diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Weilei Yao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jun Xia
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Tongxin Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
45
|
Osataphan S, Macchi C, Singhal G, Chimene-Weiss J, Sales V, Kozuka C, Dreyfuss JM, Pan H, Tangcharoenpaisan Y, Morningstar J, Gerszten R, Patti ME. SGLT2 inhibition reprograms systemic metabolism via FGF21-dependent and -independent mechanisms. JCI Insight 2019; 4:123130. [PMID: 30843877 DOI: 10.1172/jci.insight.123130] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022] Open
Abstract
Pharmacologic inhibition of the renal sodium/glucose cotransporter-2 induces glycosuria and reduces glycemia. Given that SGLT2 inhibitors (SGLT2i) reduce mortality and cardiovascular risk in type 2 diabetes, improved understanding of molecular mechanisms mediating these metabolic effects is required. Treatment of obese but nondiabetic mice with the SGLT2i canagliflozin (CANA) reduces adiposity, improves glucose tolerance despite reduced plasma insulin, increases plasma ketones, and improves plasma lipid profiles. Utilizing an integrated transcriptomic-metabolomics approach, we demonstrate that CANA modulates key nutrient-sensing pathways, with activation of 5' AMP-activated protein kinase (AMPK) and inhibition of mechanistic target of rapamycin (mTOR), independent of insulin or glucagon sensitivity or signaling. Moreover, CANA induces transcriptional reprogramming to activate catabolic pathways, increase fatty acid oxidation, reduce hepatic steatosis and diacylglycerol content, and increase hepatic and plasma levels of FGF21. Given that these phenotypes mirror the effects of FGF21 to promote lipid oxidation, ketogenesis, and reduction in adiposity, we hypothesized that FGF21 is required for CANA action. Using FGF21-null mice, we demonstrate that FGF21 is not required for SGLT2i-mediated induction of lipid oxidation and ketogenesis but is required for reduction in fat mass and activation of lipolysis. Taken together, these data demonstrate that SGLT2 inhibition triggers a fasting-like transcriptional and metabolic paradigm but requires FGF21 for reduction in adiposity.
Collapse
Affiliation(s)
- Soravis Osataphan
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Srinakharinwirot University, Bangkok, Thailand
| | - Chiara Macchi
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Garima Singhal
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Endocrinology and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jeremy Chimene-Weiss
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Vicencia Sales
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Chisayo Kozuka
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan M Dreyfuss
- Harvard Medical School, Boston, Massachusetts, USA.,Bioinformatics and Biostatistics Core, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Hui Pan
- Harvard Medical School, Boston, Massachusetts, USA.,Bioinformatics and Biostatistics Core, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Yanin Tangcharoenpaisan
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Jordan Morningstar
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Robert Gerszten
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Mary-Elizabeth Patti
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Defective fasting-induced PKA activation impairs adipose tissue glycogen degradation in obese Zucker rats. Int J Obes (Lond) 2019; 44:500-509. [PMID: 30705392 DOI: 10.1038/s41366-019-0327-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/20/2018] [Accepted: 01/11/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Obesity is associated with development of insulin resistance in adipose tissue (AT). Human obesity has been associated with increased glycogen deposition in adipocytes. Adipocytes synthesise glycogen prior to the formation of lipids. The present study examined adipose glycogen content in obese Zucker rats and the effect of fasting on glycogen-metabolising enzymes. We hypothesised that obesity imposes a blunted response to fasting through impaired activation of glycogen-metabolizing enzymes, which dampens glycogen mobilization in obese Zucker rats. METHODS We investigated the effect of 24h fasting on AT glycogen metabolism in 12-week old obese Zucker rats. Epididymal fat pads were collected from rats fed ad-libitum and fasted for 24h. Glycogen content, glycogen synthase and phosphorylase enzyme activity, and PKA activity were analysed as well as total and phosphorylated protein content for glycogen-metabolizing enzymes glycogen synthase and phosphorylase, glucose transporter GLUT4, and cAMP-dependent response element binding protein levels. RESULTS Twelve-week old obese Zucker rats showed increased AT glycogen content (adipose glycogen content [mean ± SD], lean: 3.95 ± 2.78 to 0.75 + 0.69 µg.mg-1; p < 0.005 fed vs fasted, and obese: 5.23 ± 3.38 to 5.019 ± 1.99 µg.mg-1; p = ns fed and fasted and p < 0.005 lean vs obese), and impaired fasting-induced glycogen mobilization following a 24h fast. These defects were associated with dysfunctional glycogen-metabolizing enzymes, characterized by: (1) blunted phosphorylation-mediated activation and downregulated protein expression of glycogen phosphorylase, and (2) an impaired phosphorylation-mediated inactivation of glycogen synthase. Furthermore, these defects were related to impaired fasting-induced protein kinase A (PKA) activation. CONCLUSION This study provides evidence of a defective glycogen metabolism in the adipose associated with impaired fasting-induced activation of the upstream kinase protein kinase A, which render a converging point to obesity-related primary alterations in carbohydrate and lipid metabolism in the AT.
Collapse
|
47
|
Wei J, Chen L, Li F, Yuan Y, Wang Y, Xia W, Zhang Y, Xu Y, Yang Z, Gao B, Jin C, Melo-Cardenas J, Green RM, Pan H, Wang J, He F, Zhang K, Fang D. HRD1-ERAD controls production of the hepatokine FGF21 through CREBH polyubiquitination. EMBO J 2018; 37:embj.201898942. [PMID: 30389664 DOI: 10.15252/embj.201898942] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 09/03/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum-associated protein degradation (ERAD) is responsible for recognizing and retro-translocating protein substrates, misfolded or not, from the ER for cytosolic proteasomal degradation. HMG-CoA Reductase (HMGCR) Degradation protein-HRD1-was initially identified as an E3 ligase critical for ERAD. However, its physiological functions remain largely undefined. Herein, we discovered that hepatic HRD1 expression is induced in the postprandial condition upon mouse refeeding. Mice with liver-specific HRD1 deletion failed to repress FGF21 production in serum and liver even in the refeeding condition and phenocopy the FGF21 gain-of-function mice showing growth retardation, female infertility, and diurnal circadian behavior disruption. HRD1-ERAD facilitates the degradation of the liver-specific ER-tethered transcription factor CREBH to downregulate FGF21 expression. HRD1-ERAD catalyzes polyubiquitin conjugation onto CREBH at lysine 294 for its proteasomal degradation, bridging a multi-organ crosstalk in regulating growth, circadian behavior, and female fertility through regulating the CREBH-FGF21 regulatory axis.
Collapse
Affiliation(s)
- Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lu Chen
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Fei Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yanzhi Yuan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yajun Wang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Wanjun Xia
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuehui Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yuanming Xu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhao Yang
- Department of Immunology, Microbiology, and Biochemistry, Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chaozhi Jin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Johanna Melo-Cardenas
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Richard M Green
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hui Pan
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Kezhong Zhang
- Department of Immunology, Microbiology, and Biochemistry, Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA .,Department of Pharmacology, Dalian Medical University School of Pharmacy, Dalian, China
| |
Collapse
|
48
|
Kim T, Holleman CL, Nason S, Arble DM, Ottaway N, Chabenne J, Loyd C, Kim JA, Sandoval D, Drucker DJ, DiMarchi R, Perez-Tilve D, Habegger KM. Hepatic Glucagon Receptor Signaling Enhances Insulin-Stimulated Glucose Disposal in Rodents. Diabetes 2018; 67:2157-2166. [PMID: 30150304 PMCID: PMC6198333 DOI: 10.2337/db18-0068] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022]
Abstract
Glucagon receptor (GCGR) agonists cause hyperglycemia but also weight loss. However, GCG-like peptide 1 receptor (GLP1R)/GCGR mixed agonists do not exhibit the diabetogenic effects often attributed to GCGR activity. Thus, we sought to investigate the effect of glucagon agonism on insulin action and glucose homeostasis. Acute GCGR agonism induced immediate hyperglycemia, followed by improved glucose tolerance and enhanced glucose-stimulated insulin secretion. Moreover, acute GCGR agonism improved insulin tolerance in a dose-dependent manner in both lean and obese mice. Improved insulin tolerance was independent of GLP1R, FGF21, and hepatic glycogenolysis. Moreover, we observed increased glucose infusion rate, disposal, uptake, and suppressed endogenous glucose production during euglycemic clamps. Mice treated with insulin and GCGR agonist had enhanced phosphorylation of hepatic AKT at Ser473; this effect was reproduced in isolated mouse primary hepatocytes and resulted in increased AKT kinase activity. These data reveal that GCGR agonism enhances glucose tolerance, in part, by augmenting insulin action, with implications for the use of GCGR agonism in therapeutic strategies for diabetes.
Collapse
Affiliation(s)
- Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Cassie L Holleman
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Shelly Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Deanna M Arble
- Department of Biological Sciences, Marquette University, Milwaukee, WI
| | - Nickki Ottaway
- Metabolic Diseases Institute and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, OH
| | | | - Christine Loyd
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jeong-A Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Richard DiMarchi
- Novo Nordisk Research Center, Indianapolis, IN
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Diego Perez-Tilve
- Metabolic Diseases Institute and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Cincinnati, Cincinnati, OH
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
49
|
Chiba Y, Yamada T, Katagiri H. [Dapagliflozin, a Sodium-Glucose Co-transporter-2 Inhibitor, Acutely Reduces Energy Expenditure in Brown Adipose Tissue via Neural Signals in Mice]. YAKUGAKU ZASSHI 2018; 138:945-954. [PMID: 29962474 DOI: 10.1248/yakushi.17-00223-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Selective sodium glucose transporter-2 inhibitor (SGLT2i) treatment promotes urinary glucose excretion, thereby reducing blood glucose as well as body weight. However, only limited body weight reductions are achieved with SGLT2i administration. Hyperphagia is reportedly one of the causes of this limited weight loss. However, the effects of SGLT2i on systemic energy expenditure have not been fully elucidated. We investigated the acute effects of dapagliflozin, an SGLT2i, on systemic energy expenditure in mice. Eighteen hours after dapagliflozin administration, oxygen consumption and brown adipose tissue (BAT) expression of ucp1, a thermogenesis-related gene, were significantly decreased as compared with those after vehicle administration. In addition, dapagliflozin significantly suppressed norepinephrine (NE) turnover in BAT and c-fos expression in the rostral raphe pallidus nucleus (rRPa), which contains the sympathetic premotor neurons responsible for thermogenesis. These findings indicate that the dapagliflozin-mediated acute decrease in energy expenditure involves a reduction in BAT thermogenesis via decreased sympathetic nerve activity from the rRPa. Furthermore, common hepatic branch vagotomy abolished the reductions in ucp1 expression, NE contents in BAT, and c-fos expression in the rRPa. In addition, alterations in hepatic carbohydrate metabolism, such as decreases in glycogen contents and upregulation of phosphoenolpyruvate carboxykinase, occurred prior to the suppression of BAT thermogenesis, e.g., 6 h after dapagliflozin treatment. Collectively, these results suggest that SGLT2i acutely suppresses energy expenditure in BAT via regulation of an interorgan neural network consisting of the common hepatic vagal branch and sympathetic nerves.
Collapse
Affiliation(s)
- Yumiko Chiba
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine
| |
Collapse
|
50
|
Organic cation transporter 1 (OCT1) modulates multiple cardiometabolic traits through effects on hepatic thiamine content. PLoS Biol 2018; 16:e2002907. [PMID: 29659562 PMCID: PMC5919692 DOI: 10.1371/journal.pbio.2002907] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 04/26/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose–fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease. The liver is the major organ for glucose and lipid metabolism; impairment in liver energy metabolism is often found in metabolic disorders. Traditionally, excesses in macronutrients (fat and glucose) are linked to the development of metabolic disorders. Our study provides evidence that imbalances in a micronutrient, vitamin B1 (thiamine), can serve as an etiological cause of lipid and glucose disorders and implicates the organic cation transporter, OCT1, in these disorders. OCT1 is a key determinant of thiamine levels in the liver. In humans, reduced-function polymorphisms of OCT1 significantly associate with high LDL cholesterol levels. Using Oct1 knockout mice, we show that reduced OCT1-mediated thiamine uptake in the liver leads to reduced levels of TPP—the active metabolite of thiamine—and decreased activity of key TPP-dependent enzymes. As a result, a shift from glucose to fatty acid oxidation occurs, leading to imbalances in key metabolic intermediates, alterations in metabolic flux pathways, and disruptions of various metabolic regulatory mechanisms. The extensive characterization of Oct1 knockout mice provides evidence for the molecular mechanisms responsible for various metabolic traits and indicates an important role for imbalances in micronutrients in cardiometabolic disorders.
Collapse
|