1
|
Chen Y, Zhang Y, Jin X, Hong S, Tian H. Exerkines: Benign adaptation for exercise and benefits for non-alcoholic fatty liver disease. Biochem Biophys Res Commun 2024; 726:150305. [PMID: 38917635 DOI: 10.1016/j.bbrc.2024.150305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
Exercise has multiple beneficial effects on human metabolic health and is regarded as a "polypill" for various diseases. At present, the lack of physical activity usually causes an epidemic of chronic metabolic syndromes, including obesity, cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Remarkably, NAFLD is emerging as a serious public health issue and is associated with the development of cirrhosis and hepatocellular carcinoma. Unfortunately, specific drug therapies for NAFLD and its more severe form, non-alcoholic steatohepatitis (NASH), are currently unavailable. Lifestyle modification is the foundation of treatment recommendations for NAFLD and NASH, especially for exercise. There are under-appreciated organs that crosstalk to the liver during exercise such as muscle-liver crosstalk. Previous studies have reported that certain exerkines, such as FGF21, GDF15, irisin, and adiponectin, are beneficial for liver metabolism and have the potential to be targeted for NAFLD treatment. In addition, some of exerkines can be modified for the new proteins and get enhanced functions, like IL-6/IC7Fc. Another importance of exercise is the physiological adaptation that combats metabolic diseases. Thus, this review aims to summarize the known exerkines and utilize a multi-omics mining tool to identify more exerkines for the future research. Overall, understanding the mechanisms by which exercise-induced exerkines exert their beneficial effects on metabolic health holds promise for the development of novel therapeutic strategies for NAFLD and related diseases.
Collapse
Affiliation(s)
- Yang Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yan Zhang
- Clinical Laboratory, Suzhou Yong Ding Hospital, Suzhou, 215200, China
| | - Xingsheng Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200032, China.
| | - Haili Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
2
|
Li S, Zou T, Chen J, Li J, You J. Fibroblast growth factor 21: An emerging pleiotropic regulator of lipid metabolism and the metabolic network. Genes Dis 2024; 11:101064. [PMID: 38292170 PMCID: PMC10825286 DOI: 10.1016/j.gendis.2023.06.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/20/2023] [Accepted: 06/27/2023] [Indexed: 02/01/2024] Open
Abstract
Fibroblast growth factor 21 (FGF21) was originally identified as an important metabolic regulator which plays a crucial physiological role in regulating a variety of metabolic parameters through the metabolic network. As a novel multifunctional endocrine growth factor, the role of FGF21 in the metabolic network warrants extensive exploration. This insight was obtained from the observation that the FGF21-dependent mechanism that regulates lipid metabolism, glycogen transformation, and biological effectiveness occurs through the coordinated participation of the liver, adipose tissue, central nervous system, and sympathetic nerves. This review focuses on the role of FGF21-uncoupling protein 1 (UCP1) signaling in lipid metabolism and how FGF21 alleviates non-alcoholic fatty liver disease (NAFLD). Additionally, this review reveals the mechanism by which FGF21 governs glucolipid metabolism. Recent research on the role of FGF21 in the metabolic network has mostly focused on the crucial pathway of glucolipid metabolism. FGF21 has been shown to have multiple regulatory roles in the metabolic network. Since an adequate understanding of the concrete regulatory pathways of FGF21 in the metabolic network has not been attained, this review sheds new light on the metabolic mechanisms of FGF21, explores how FGF21 engages different tissues and organs, and lays a theoretical foundation for future in-depth research on FGF21-targeted treatment of metabolic diseases.
Collapse
Affiliation(s)
| | | | - Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jiaming Li
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| |
Collapse
|
3
|
Marinescu SC(N, Apetroaei MM, Nedea MI(I, Arsene AL, Velescu BȘ, Hîncu S, Stancu E, Pop AL, Drăgănescu D, Udeanu DI. Dietary Influence on Drug Efficacy: A Comprehensive Review of Ketogenic Diet-Pharmacotherapy Interactions. Nutrients 2024; 16:1213. [PMID: 38674903 PMCID: PMC11054576 DOI: 10.3390/nu16081213] [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: 03/28/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
It is widely acknowledged that the ketogenic diet (KD) has positive physiological effects as well as therapeutic benefits, particularly in the treatment of chronic diseases. Maintaining nutritional ketosis is of utmost importance in the KD, as it provides numerous health advantages such as an enhanced lipid profile, heightened insulin sensitivity, decreased blood glucose levels, and the modulation of diverse neurotransmitters. Nevertheless, the integration of the KD with pharmacotherapeutic regimens necessitates careful consideration. Due to changes in their absorption, distribution, metabolism, or elimination, the KD can impact the pharmacokinetics of various medications, including anti-diabetic, anti-epileptic, and cardiovascular drugs. Furthermore, the KD, which is characterised by the intake of meals rich in fats, has the potential to impact the pharmacokinetics of specific medications with high lipophilicity, hence enhancing their absorption and bioavailability. However, the pharmacodynamic aspects of the KD, in conjunction with various pharmaceutical interventions, can provide either advantageous or detrimental synergistic outcomes. Therefore, it is important to consider the pharmacokinetic and pharmacodynamic interactions that may arise between the KD and various drugs. This assessment is essential not only for ensuring patients' compliance with treatment but also for optimising the overall therapeutic outcome, particularly by mitigating adverse reactions. This highlights the significance and necessity of tailoring pharmacological and dietetic therapies in order to enhance the effectiveness and safety of this comprehensive approach to managing chronic diseases.
Collapse
Affiliation(s)
- Simona Cristina (Nicolescu) Marinescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
- Amethyst Radiotherapy Center, 42, Drumul Odăi, 075100 Otopeni, Romania
| | - Miruna-Maria Apetroaei
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
| | - Marina Ionela (Ilie) Nedea
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
| | - Andreea Letiția Arsene
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
- Marius Nasta Institute of Pneumophthiology, 90, Viilor Street, 050159 Bucharest, Romania
| | - Bruno Ștefan Velescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
| | - Sorina Hîncu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
- Fundeni Clinical Institute, 258, Fundeni Street, 022328 Bucharest, Romania
| | - Emilia Stancu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
| | - Anca Lucia Pop
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
| | - Doina Drăgănescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
| | - Denisa Ioana Udeanu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6, Traian Vuia Street, 020956 Bucharest, Romania (A.L.A.); (B.Ș.V.); (S.H.); (E.S.); (A.L.P.); (D.D.); (D.I.U.)
- Marius Nasta Institute of Pneumophthiology, 90, Viilor Street, 050159 Bucharest, Romania
| |
Collapse
|
4
|
Kukułowicz J, Pietrzak-Lichwa K, Klimończyk K, Idlin N, Bajda M. The SLC6A15-SLC6A20 Neutral Amino Acid Transporter Subfamily: Functions, Diseases, and Their Therapeutic Relevance. Pharmacol Rev 2023; 76:142-193. [PMID: 37940347 DOI: 10.1124/pharmrev.123.000886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/07/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023] Open
Abstract
The neutral amino acid transporter subfamily that consists of six members, consecutively SLC6A15-SLC620, also called orphan transporters, represents membrane, sodium-dependent symporter proteins that belong to the family of solute carrier 6 (SLC6). Primarily, they mediate the transport of neutral amino acids from the extracellular milieu toward cell or storage vesicles utilizing an electric membrane potential as the driving force. Orphan transporters are widely distributed throughout the body, covering many systems; for instance, the central nervous, renal, or intestinal system, supplying cells into molecules used in biochemical, signaling, and building pathways afterward. They are responsible for intestinal absorption and renal reabsorption of amino acids. In the central nervous system, orphan transporters constitute a significant medium for the provision of neurotransmitter precursors. Diseases related with aforementioned transporters highlight their significance; SLC6A19 mutations are associated with metabolic Hartnup disorder, whereas altered expression of SLC6A15 has been associated with a depression/stress-related disorders. Mutations of SLC6A18-SLCA20 cause iminoglycinuria and/or hyperglycinuria. SLC6A18-SLC6A20 to reach the cellular membrane require an ancillary unit ACE2 that is a molecular target for the spike protein of the SARS-CoV-2 virus. SLC6A19 has been proposed as a molecular target for the treatment of metabolic disorders resembling gastric surgery bypass. Inhibition of SLC6A15 appears to have a promising outcome in the treatment of psychiatric disorders. SLC6A19 and SLC6A20 have been suggested as potential targets in the treatment of COVID-19. In this review, we gathered recent advances on orphan transporters, their structure, functions, related disorders, and diseases, and in particular their relevance as therapeutic targets. SIGNIFICANCE STATEMENT: The following review systematizes current knowledge about the SLC6A15-SLCA20 neutral amino acid transporter subfamily and their therapeutic relevance in the treatment of different diseases.
Collapse
Affiliation(s)
- Jędrzej Kukułowicz
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Pietrzak-Lichwa
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Klimończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Nathalie Idlin
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
5
|
Bahijri S, Eldakhakhny B, Enani S, Ajabnoor G, Al-Mowallad AS, Alsheikh L, Alhozali A, Alamoudi AA, Borai A, Tuomilehto J. Fibroblast Growth Factor 21: A More Effective Biomarker Than Free Fatty Acids and Other Insulin Sensitivity Measures for Predicting Non-alcoholic Fatty Liver Disease in Saudi Arabian Type 2 Diabetes Patients. Cureus 2023; 15:e50524. [PMID: 38222178 PMCID: PMC10787595 DOI: 10.7759/cureus.50524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2023] [Indexed: 01/16/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is more prevalent among individuals with type 2 diabetes (T2DM), elevating their risk of cardiovascular diseases (CVDs) and premature mortality. There is a need to modify treatment strategies to prevent or delay these adverse outcomes. Currently, there are no sensitive or specific biomarkers for predicting NAFLD in Saudi T2DM patients. Therefore, we aimed to explore the possibility of using fibroblast growth factor 21 (FGF-21), free fatty acids (FFAs), homeostatic model assessment for insulin resistance (HOMA-IR), and quantitative insulin sensitivity check index (QUICKI) as possible markers. Methodology In this study, a total of 67 T2DM patients were recruited. NAFLD was detected by ultrasonography in 28 patients. Plasma glucose, FFAs, FGF-21, and serum insulin were measured in fasting blood samples. HOMA-IR and QUICKI were calculated. The means of the two groups with and without NAFLD were statistically compared. The receiver operating characteristics (ROC) curve and the area under the curve (AUC) were used to assess the ability to identify NAFLD. Results The mean levels of FGF-21 and HOMA-IR were significantly higher and that of QUICKI was significantly lower in patients with NAFLD than in those without (p < 0.001, p = 0.023, and p = 0.018, respectively). FGF-21 had the highest AUC to identify NAFLD (AUC = 0.981, 95% confidence interval = 0.954-1, P < 0.001). The AUCs for HOMA-IR, QUICKI, and FFA were <0.7. The highest sensitivity, specificity, positive likelihood ratio, and the lowest negative likelihood ratio were found when FGF-21 was used to predict NAFLD. Conclusions FGF-21 may be used as a biomarker to predict NAFLD in people with T2DM due to its high sensitivity and specificity compared to the other markers.
Collapse
Affiliation(s)
- Suhad Bahijri
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Basmah Eldakhakhny
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Sumia Enani
- Department of Food and Nutrition, Faculty of Human Sciences and Design, King Abdulaziz University, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Ghada Ajabnoor
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, SAU
| | - Alaa S Al-Mowallad
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
| | - Lubna Alsheikh
- Department of Biochemistry, King Abdulaziz University, Jeddah, SAU
| | - Amani Alhozali
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah, SAU
| | - Aliaa A Alamoudi
- Department of Clinical Biochemistry, King Abdulaziz University Faculty of Medicine, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
| | - Anwar Borai
- King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Saudi Diabetes Research Group, Deanship of Scientific Research, King Abdulaziz University, Jeddah, SAU
| | - Jaakko Tuomilehto
- Department of Public Health, University of Helsinki, Helsinki, FIN
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, FIN
| |
Collapse
|
6
|
Falkevall A, Mehlem A, Folestad E, Ning FC, Osorio-Conles Ó, Radmann R, de Hollanda A, Wright SD, Scotney P, Nash A, Eriksson U. Inhibition of VEGF-B signaling prevents non-alcoholic fatty liver disease development by targeting lipolysis in the white adipose tissue. J Hepatol 2023; 78:901-913. [PMID: 36717026 DOI: 10.1016/j.jhep.2023.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/22/2022] [Accepted: 01/13/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND & AIMS Hepatic steatosis is a hallmark of non-alcoholic fatty liver disease (NAFLD), a common comorbidity in type 2 diabetes mellitus (T2DM). The pathogenesis of NAFLD is complex and involves the crosstalk between the liver and the white adipose tissue (WAT). Vascular endothelial growth factor B (VEGF-B) has been shown to control tissue lipid accumulation by regulating the transport properties of the vasculature. The role of VEGF-B signaling and the contribution to hepatic steatosis and NAFLD in T2DM is currently not understood. METHODS C57BL/6 J mice treated with a neutralizing antibody against VEGF-B, or mice with adipocyte-specific overexpression or under-expression of VEGF-B (AdipoqCre+/VEGF-BTG/+ mice and AdipoqCre+/Vegfbfl/+mice) were subjected to a 6-month high-fat diet (HFD), or chow-diet, whereafter NAFLD development was assessed. VEGF-B expression was analysed in WAT biopsies from patients with obesity and NAFLD in a pre-existing clinical cohort (n = 24 patients with NAFLD and n = 24 without NAFLD) and correlated to clinicopathological features. RESULTS Pharmacological inhibition of VEGF-B signaling in diabetic mice reduced hepatic steatosis and NAFLD by blocking WAT lipolysis. Mechanistically we show, by using HFD-fed AdipoqCre+/VEGF-BTG/+ mice and HFD-fed AdipoqCre+/Vegfbfl/+mice, that inhibition of VEGF-B signaling targets lipolysis in adipocytes. Reducing VEGF-B signaling ameliorated NAFLD by decreasing WAT inflammation, resolving WAT insulin resistance, and lowering the activity of the hormone sensitive lipase. Analyses of human WAT biopsies from individuals with NAFLD provided evidence supporting the contribution of VEGF-B signaling to NAFLD development. VEGF-B expression levels in adipocytes from two WAT depots correlated with development of dysfunctional WAT and NAFLD in humans. CONCLUSIONS Taken together, our data from mouse models and humans suggest that VEGF-B antagonism may represent an approach to combat NAFLD by targeting hepatic steatosis through suppression of lipolysis. IMPACT AND IMPLICATIONS Non-alcoholic fatty liver disease (NAFLD) is a common comorbidity in type 2 diabetes mellitus (T2DM) and has a global prevalence of between 25-29%. There are currently no approved drugs for NAFLD, and given the scale of the ongoing diabetes epidemics, there is an urgent need to identify new treatment options. Our work suggests that VEGF-B antagonism may represent an approach to combat NAFLD by targeting hepatic steatosis through suppression of lipolysis. The neutralizing anti-VEGF-B antibody, which was used in this study, has already entered clinical trials for patients with diabetes. Therefore, we believe that our results are of great general interest to a broad audience, including patients and patient organizations, the medical community, academia, the life science industry and the public.
Collapse
Affiliation(s)
- Annelie Falkevall
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Annika Mehlem
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Erika Folestad
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Frank Chenfei Ning
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Óscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Rosa Radmann
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ana de Hollanda
- Obesity Unit. Clinical Hospital of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | | | | | - Andrew Nash
- CSL Innovation Pty Ltd, Parkville, Victoria, Australia
| | - Ulf Eriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
7
|
Yaghoubi F, Darand M, Vasmehjani AA, Darabi Z, Talenezhad N, Mirzavandi F, Hosseinzadeh M. Adherence to low carbohydrate diets and non-alcoholic fatty liver disease: a case control study. BMC Nutr 2022; 8:140. [PMID: 36447244 PMCID: PMC9706826 DOI: 10.1186/s40795-022-00625-5] [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: 06/29/2021] [Accepted: 10/26/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is defined as the excessive accumulation of fat in the liver cells of people who do not drink alcohol. The aim of study is investigated the association between low carbohydrate diets (LCDs) and NAFLD. METHODS This age and gender-matched case-control study was conducted on 120 patients newly diagnosed with NAFLD and 120 adults without NAFLD. Diagnosis of NAFLD based on laboratory tests and abdominal ultrasound. Low carbohydrate diets score calculated on the percentage of energy as carbohydrate, fat, and protein. Participants in the highest rank intake of fat and protein and lowest intake of carbohydrate received 10 points. Multivariable logistic odds ratio was used for examine the relation between LCDs and NAFLD. RESULTS This study showed subjects in the highest tertile of LCD has more intake of zinc and vitamin B12 compare to lowest. Also, intake of protein (p = 0.02) carbohydrate (p < 0.02) and cholesterol (p = 0.02) were significantly higher in patient with NAFLD compare to control subjects. There was no significant association between LCD and risk of NAFLD (OR: 1.36; 95% CI: 0.97-1.92; P-trend = 0.13) in crude and adjusted (OR: 1.31; 95% CI: 0.84-2.04; P-trend = 0.23) model. CONCLUSION However, we showed that intake of protein, carbohydrate and cholesterol are higher in NAFLD, but our results of study showed that LCDs with higher proportion intakes of protein and fat was not associated with NAFLD. Further prospective studies are required for confirm these associations.
Collapse
Affiliation(s)
- Fatemeh Yaghoubi
- grid.412505.70000 0004 0612 5912Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran ,grid.412505.70000 0004 0612 5912Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mina Darand
- grid.411036.10000 0001 1498 685XDepartment of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aazam Ahmadi Vasmehjani
- grid.412505.70000 0004 0612 5912Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran ,Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Zahra Darabi
- grid.412505.70000 0004 0612 5912Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran ,Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Nasir Talenezhad
- grid.412505.70000 0004 0612 5912Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran ,Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Farhang Mirzavandi
- grid.412505.70000 0004 0612 5912Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran ,Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Hosseinzadeh
- grid.412505.70000 0004 0612 5912Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran ,Department of Nutrition, School of Public Health, Shahid Sadughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
8
|
Power Guerra N, Leyens K, Müller L, Brauer D, Janowitz D, Schlick S, Pilz K, Grabe HJ, Vollmar B, Kuhla A. The effect of different weight loss strategies to treat non-alcoholic fatty liver disease focusing on fibroblast growth factor 21. Front Nutr 2022; 9:935805. [PMID: 36034917 PMCID: PMC9399780 DOI: 10.3389/fnut.2022.935805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Obesity, often associated with non-alcoholic fatty liver disease (NAFLD), is characterized by an imbalance between energy expenditure and food intake, which is also reflected by desensitization of fibroblast growth factor 21 (FGF21). FGF21 is strongly influenced, among others, by TNFα, which is known to be upregulated in obesity-induced inflammation. Successful long-term treatments of NAFLD might be dietary modification, exercise, or fasting. Materials and methods Whether succeeded NAFLD recovery is linked with improved FGF21 sensitivity and finally reverted FGF21 resistance was the focus of the present study. For this purpose, mice received a high-fat diet (HFD) for 6 months to establish obesity. Afterward, the mice were subjected to three different weight loss interventions, namely, dietary change to low-fat diet (LFD), treadmill training, and/or time-restricted feeding for additional 6 months, whereas one group remained on HFD. Results In addition to the expected decrease in NAFLD activity with dietary change, this was also observed in the HFD group with additional time-restricted feeding. There was also an associated decrease in hepatic TNFα and FGF21 expression and an increase in ß-klotho expression, demonstrated mainly by using principal component analysis. Pearson correlation analysis shows that independent of any intervention, TNFα expression decreased with improved NAFLD recovery. This was accompanied with higher FGF21 sensitivity, as expressed by an increase in β-klotho and FGFR1c expression and concomitantly decreased FGF21 levels. Conclusion In summary, we conclude that successful NAFLD therapy is associated with a reversion of the TNFα-triggered FGF21-resistant state or desensitization.
Collapse
Affiliation(s)
- Nicole Power Guerra
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany.,Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Katharina Leyens
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Luisa Müller
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany.,Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - David Brauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Deborah Janowitz
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany.,Clinic for Psychiatry and Psychotherapy, HELIOS Hanseklinikum Stralsund, Stralsund, Germany
| | - Samin Schlick
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany.,Clinic for Psychiatry and Psychotherapy, HELIOS Hanseklinikum Stralsund, Stralsund, Germany
| | - Kristin Pilz
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry, University Medicine Greifswald, Greifswald, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
9
|
Huang Q, Wu M, Wu X, Zhang Y, Xia Y. Muscle-to-tumor crosstalk: The effect of exercise-induced myokine on cancer progression. Biochim Biophys Acta Rev Cancer 2022; 1877:188761. [PMID: 35850277 DOI: 10.1016/j.bbcan.2022.188761] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 02/07/2023]
Abstract
Physical exercise has gradually become a focus in cancer treatment due to its pronounced role in reducing cancer risk, enhancing therapeutic efficacy, and improving prognosis. In recent decades, skeletal muscles have been considered endocrine organs, exerting their biological functions via the endocrine, autocrine, and paracrine systems by secreting various types of myokines. The amount of myokines secreted varies depending on the intensity, type, and duration of exercise. Recent studies have shown that muscle-derived myokines are highly involved the effects of exercise on cancer. Multiple myokines, such as interleukin-6 (IL-6), oncostatin M (OSM), secreted protein acidic and rich in cysteine (SPARC), and irisin, directly mediate cancer progression by influencing the proliferation, apoptosis, stemness, drug resistance, metabolic reprogramming, and epithelial-mesenchymal transformation (EMT) of cancer cells. In addition, IL-6, interleukin-8 (IL-8), interleukin-15 (IL-15), brain-derived neurotrophic factor (BDNF), and irisin can improve obesity-induced inflammation by stimulating lipolysis of adipose tissues, promoting glucose uptake, and accelerating the browning of white fat. Furthermore, some myokines could regulate the tumor microenvironment, such as angiogenesis and the immune microenvironment. Cancer cachexia occurs in up to 80% of cancer patients and is responsible for 22%-30% of patient deaths. It is characterized by systemic inflammation and decreased muscle mass. Exercise-induced myokine production is important in regulating cancer cachexia. This review summarizes the roles and underlying mechanisms of myokines, such as IL-6, myostatin, IL-15, irisin, fibroblast growth factor 21 (FGF21) and musclin, in cancer cachexia. Through comprehensive analysis, we conclude that myokines are potential targets for inhibiting cancer progression and the associated cachexia.
Collapse
Affiliation(s)
- Qianrui Huang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengling Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu 610041, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yong Xia
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu 610041, China.
| |
Collapse
|
10
|
Norwitz NG, Soto-Mota A, Kaplan B, Ludwig DS, Budoff M, Kontush A, Feldman D. The Lipid Energy Model: Reimagining Lipoprotein Function in the Context of Carbohydrate-Restricted Diets. Metabolites 2022; 12:metabo12050460. [PMID: 35629964 PMCID: PMC9147253 DOI: 10.3390/metabo12050460] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
When lean people adopt carbohydrate-restricted diets (CRDs), they may develop a lipid profile consisting of elevated LDL-cholesterol (LDL-C) and HDL-cholesterol (HDL-C) with low triglycerides (TGs). The magnitude of this lipid profile correlates with BMI such that those with lower BMI exhibit larger increases in both LDL-C and HDL-C. The inverse association between BMI and LDL-C and HDL-C change on CRD contributed to the discovery of a subset of individuals—termed Lean Mass Hyper-Responders (LMHR)—who, despite normal pre-diet LDL-C, as compared to non-LMHR (mean levels of 148 and 145 mg/dL, respectively), exhibited a pronounced hyperlipidemic response to a CRD, with mean LDL-C and HDL-C levels increasing to 320 and 99 mg/dL, respectively, in the context of mean TG of 47 mg/dL. In some LMHR, LDL-C levels may be in excess of 500 mg/dL, again, with relatively normal pre-diet LDL-C and absent of genetic findings indicative of familial hypercholesterolemia in those who have been tested. The Lipid Energy Model (LEM) attempts to explain this metabolic phenomenon by positing that, with carbohydrate restriction in lean persons, the increased dependence on fat as a metabolic substrate drives increased hepatic secretion and peripheral uptake of TG contained within very low-density lipoproteins (VLDL) by lipoprotein lipase, resulting in marked elevations of LDL-C and HDL-C, and low TG. Herein, we review the core features of the LEM. We review several existing lines of evidence supporting the model and suggest ways to test the model’s predictions.
Collapse
Affiliation(s)
- Nicholas G. Norwitz
- Harvard Medical School, Boston, MA 02115, USA;
- Correspondence: (N.G.N.); (D.F.)
| | - Adrian Soto-Mota
- Metabolic Diseases Research Unit, National Institute for Medical Sciences and Nutrition Salvador Zubiran, Tlalpan, CDMX 14080, Mexico;
| | - Bob Kaplan
- Citizen Science Foundation, Las Vegas, NV 89139, USA;
| | - David S. Ludwig
- Harvard Medical School, Boston, MA 02115, USA;
- New Balance Foundation Obesity Prevention Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Matthew Budoff
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
| | - Anatol Kontush
- National Institute for Health and Medical Research (INSERM), UMRS 1166 ICAN, Faculty of Medicine Pitié-Salpêtrière, Sorbonne University, 75013 Paris, France;
| | - David Feldman
- Citizen Science Foundation, Las Vegas, NV 89139, USA;
- Correspondence: (N.G.N.); (D.F.)
| |
Collapse
|
11
|
Saito H, Wada N, Iida K. Isonitrogenous low-carbohydrate diet elicits specific changes in metabolic gene expression in the skeletal muscle of exercise-trained mice. PLoS One 2022; 17:e0262875. [PMID: 35061842 PMCID: PMC8782354 DOI: 10.1371/journal.pone.0262875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 01/06/2022] [Indexed: 11/28/2022] Open
Abstract
With the renewed interest in low-carbohydrate diets (LCDs) in the sports field, a few animal studies have investigated their potential. However, most rodent studies have used an LCD containing low protein, which does not recapitulate a human LCD, and the muscle-specific adaptation in response to an LCD remains unclear. Therefore, we investigated the effects of two types of LCDs, both containing the same proportion of protein as a regular diet (isonitrogenous LCD; INLCD), on body composition, exercise performance, and metabolic fuel selection at the genetic level in the skeletal muscles of exercise-trained mice. Three groups of mice (n = 8 in each group), one fed a regular AIN-93G diet served as the control, and the others fed either of the two INLCDs containing 20% protein and 10% carbohydrate (INLCD-10%) or 20% protein and 1% carbohydrate (INLCD-1%) had a regular exercise load (5 times/week) for 12 weeks. Body weight and muscle mass did not decrease in either of the INLCD-fed groups, and the muscle glycogen levels and endurance capacity did not differ among the three groups. Only in the mice fed INLCD-1% did the plasma ketone concentration significantly increase, and gene expression related to glucose utilization significantly declined in the muscles. Both INLCD-1% and INLCD-10% consumption increased gene expression related to lipid utilization. These results suggest that, although INLCD treatment did not affect endurance capacity, it helped maintain muscle mass and glycogen content regardless of the glucose intake restrictions in trained mice. Moreover, an INLCD containing a low carbohydrate content might present an advantage by increasing lipid oxidation without ketosis and suppressing muscle glucose utilization.
Collapse
Affiliation(s)
- Hazuki Saito
- Department of Food and Nutrition Science, Graduate School of Humanities and Sciences, Ochanomizu University, Otsuka, Bunkyo, Tokyo, Japan
| | - Naoko Wada
- Department of Food and Nutrition Science, Graduate School of Humanities and Sciences, Ochanomizu University, Otsuka, Bunkyo, Tokyo, Japan
| | - Kaoruko Iida
- Department of Food and Nutrition Science, Graduate School of Humanities and Sciences, Ochanomizu University, Otsuka, Bunkyo, Tokyo, Japan
- The Institute for Human Life Innovation, Ochanomizu University, Otsuka, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
12
|
Green CL, Pak HH, Richardson NE, Flores V, Yu D, Tomasiewicz JL, Dumas SN, Kredell K, Fan JW, Kirsh C, Chaiyakul K, Murphy ME, Babygirija R, Barrett-Wilt GA, Rabinowitz J, Ong IM, Jang C, Simcox J, Lamming DW. Sex and genetic background define the metabolic, physiologic, and molecular response to protein restriction. Cell Metab 2022; 34:209-226.e5. [PMID: 35108511 PMCID: PMC8865085 DOI: 10.1016/j.cmet.2021.12.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2021] [Accepted: 12/20/2021] [Indexed: 02/03/2023]
Abstract
Low-protein diets promote metabolic health in humans and rodents. Despite evidence that sex and genetic background are key factors in the response to diet, most protein intake studies examine only a single strain and sex of mice. Using multiple strains and both sexes of mice, we find that improvements in metabolic health in response to reduced dietary protein strongly depend on sex and strain. While some phenotypes were conserved across strains and sexes, including increased glucose tolerance and energy expenditure, we observed high variability in adiposity, insulin sensitivity, and circulating hormones. Using a multi-omics approach, we identified mega-clusters of differentially expressed hepatic genes, metabolites, and lipids associated with each phenotype, providing molecular insight into the differential response to protein restriction. Our results highlight the importance of sex and genetic background in the response to dietary protein level, and the potential importance of a personalized medicine approach to dietary interventions.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nicole E Richardson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Victoria Flores
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Deyang Yu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jay L Tomasiewicz
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Sabrina N Dumas
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Katherine Kredell
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Jesse W Fan
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Charlie Kirsh
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Krittisak Chaiyakul
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michaela E Murphy
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Joshua Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Irene M Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Cholsoon Jang
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Judith Simcox
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA; University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
13
|
Zhu H, Bi D, Zhang Y, Kong C, Du J, Wu X, Wei Q, Qin H. Ketogenic diet for human diseases: the underlying mechanisms and potential for clinical implementations. Signal Transduct Target Ther 2022; 7:11. [PMID: 35034957 PMCID: PMC8761750 DOI: 10.1038/s41392-021-00831-w] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/21/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
The ketogenic diet (KD) is a high-fat, adequate-protein, and very-low-carbohydrate diet regimen that mimics the metabolism of the fasting state to induce the production of ketone bodies. The KD has long been established as a remarkably successful dietary approach for the treatment of intractable epilepsy and has increasingly garnered research attention rapidly in the past decade, subject to emerging evidence of the promising therapeutic potential of the KD for various diseases, besides epilepsy, from obesity to malignancies. In this review, we summarize the experimental and/or clinical evidence of the efficacy and safety of the KD in different diseases, and discuss the possible mechanisms of action based on recent advances in understanding the influence of the KD at the cellular and molecular levels. We emphasize that the KD may function through multiple mechanisms, which remain to be further elucidated. The challenges and future directions for the clinical implementation of the KD in the treatment of a spectrum of diseases have been discussed. We suggest that, with encouraging evidence of therapeutic effects and increasing insights into the mechanisms of action, randomized controlled trials should be conducted to elucidate a foundation for the clinical use of the KD.
Collapse
Affiliation(s)
- Huiyuan Zhu
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Youhua Zhang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Kong
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiahao Du
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Xiawei Wu
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinical College, Anhui Medical University, Hefei, China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Spann RA, Morrison CD, den Hartigh LJ. The Nuanced Metabolic Functions of Endogenous FGF21 Depend on the Nature of the Stimulus, Tissue Source, and Experimental Model. Front Endocrinol (Lausanne) 2022; 12:802541. [PMID: 35046901 PMCID: PMC8761941 DOI: 10.3389/fendo.2021.802541] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/09/2021] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a hormone that is involved in the regulation of lipid, glucose, and energy metabolism. Pharmacological FGF21 administration promotes weight loss and improves insulin sensitivity in rodents, non-human primates, and humans. However, pharmacologic effects of FGF21 likely differ from its physiological effects. Endogenous FGF21 is produced by many cell types, including hepatocytes, white and brown adipocytes, skeletal and cardiac myocytes, and pancreatic beta cells, and acts on a diverse array of effector tissues such as the brain, white and brown adipose tissue, heart, and skeletal muscle. Different receptor expression patterns dictate FGF21 function in these target tissues, with the primary effect to coordinate responses to nutritional stress. Moreover, different nutritional stimuli tend to promote FGF21 expression from different tissues; i.e., fasting induces hepatic-derived FGF21, while feeding promotes white adipocyte-derived FGF21. Target tissue effects of FGF21 also depend on its capacity to enter the systemic circulation, which varies widely from known FGF21 tissue sources in response to various stimuli. Due to its association with obesity and non-alcoholic fatty liver disease, the metabolic effects of endogenously produced FGF21 during the pathogenesis of these conditions are not well known. In this review, we will highlight what is known about endogenous tissue-specific FGF21 expression and organ cross-talk that dictate its diverse physiological functions, with particular attention given to FGF21 responses to nutritional stress. The importance of the particular experimental design, cellular and animal models, and nutritional status in deciphering the diverse metabolic functions of endogenous FGF21 cannot be overstated.
Collapse
Affiliation(s)
- Redin A. Spann
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Christopher D. Morrison
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Laura J. den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA, United States
- Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
15
|
Diener JL, Mowbray S, Huang WJ, Yowe D, Xu J, Caplan S, Misra A, Kapur A, Shapiro J, Ke X, Wu X, Bose A, Panza D, Chen M, Beaulieu V, Gao J. FGF21 Normalizes Plasma Glucose in Mouse Models of Type 1 Diabetes and Insulin Receptor Dysfunction. Endocrinology 2021; 162:6267686. [PMID: 33951176 DOI: 10.1210/endocr/bqab092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 11/19/2022]
Abstract
Fibroblast growth factor (FGF) 21 is a member of the FGF family of proteins. The biological activity of FGF21 was first shown to induce insulin-independent glucose uptake in adipocytes through the GLUT1 transporter. Subsequently, it was shown to have effects on the liver to increase fatty acid oxidation. FGF21 treatment provides beneficial metabolic effects in both animal models and patients with obesity, type 2 diabetes mellitus (T2D) and/or fatty liver disease. In this paper, we revisited the original finding and found that insulin-independent glucose uptake in adipocytes is preserved in the presence of an insulin receptor antagonist. Using a 40-kDa PEGylated (PEG) and half-life extended form of FGF21 (FGF21-PEG), we extended these in vitro results to 2 different mouse models of diabetes. FGF21-PEG normalized plasma glucose in streptozotocin-treated mice, a model of type 1 diabetes (T1D), without restoring pancreatic β-cell function. FGF21-PEG also normalized plasma glucose levels and improved glucose tolerance in mice chronically treated with an insulin competitive insulin receptor antagonist, a model of autoimmune/type-B insulin resistance. These data extend the pharmacological potential of FGF21 beyond the settings of T2D, fatty liver, and obesity.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes/drug effects
- Adipocytes/metabolism
- Animals
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Fibroblast Growth Factors/pharmacology
- HEK293 Cells
- Humans
- Hyperglycemia/blood
- Hyperglycemia/etiology
- Hyperglycemia/pathology
- Hyperglycemia/prevention & control
- Insulin/metabolism
- Insulin Resistance/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Obese
- Obesity/blood
- Obesity/complications
- Obesity/pathology
- Receptor, Insulin/antagonists & inhibitors
- Receptor, Insulin/drug effects
- Receptor, Insulin/physiology
- Streptozocin
Collapse
Affiliation(s)
- John L Diener
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Sarah Mowbray
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Waan-Jeng Huang
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - David Yowe
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Jian Xu
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Shari Caplan
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Abhay Misra
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Ankur Kapur
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Jeffrey Shapiro
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Xiaoling Ke
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Xiaoping Wu
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Avirup Bose
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Darrell Panza
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Min Chen
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Valerie Beaulieu
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Jiaping Gao
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| |
Collapse
|
16
|
Choksomngam Y, Pattanakuhar S, Chattipakorn N, Chattipakorn SC. The metabolic role of spermidine in obesity: Evidence from cells to community. Obes Res Clin Pract 2021; 15:315-326. [PMID: 34217652 DOI: 10.1016/j.orcp.2021.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 05/15/2021] [Accepted: 06/20/2021] [Indexed: 01/07/2023]
Abstract
Spermidine is a natural polyamine existing in all living cells known to play an important role in cellular functions. Recently, several studies have reported the effect of alterations in the spermidine pool on metabolic pathways. It has been shown that activation of spermidine/spermine N-1-acetyl-transferase (SSAT), the rate-limiting enzyme in polyamine catabolism, improved glucose and lipid metabolism. In addition, spermidine supplementation has been shown to protect against diet-induced obesity in animal models. However, some clinical studies demonstrated that polyamine levels are increased in childhood obesity and metabolic syndrome patients with type 2 diabetes (T2DM), while polyamine-rich food is associated with a lower incidence of cardiovascular disease (CVD). Therefore, this review aims to summarize and discuss the evidence from in vitro, in vivo and clinical studies on the possible roles of spermidine on metabolic pathways under physiological and obese conditions. All consistent and inconsistent findings are discussed and further studies aiming to fill any gaps in the knowledge are proposed.
Collapse
Affiliation(s)
- Yanee Choksomngam
- Department of Family Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sintip Pattanakuhar
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand; Department of Rehabilitation Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
17
|
Ketogenic Diet Enhances the Cholesterol Accumulation in Liver and Augments the Severity of CCl 4 and TAA-Induced Liver Fibrosis in Mice. Int J Mol Sci 2021; 22:ijms22062934. [PMID: 33805788 PMCID: PMC7998170 DOI: 10.3390/ijms22062934] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/25/2022] Open
Abstract
Persistent chronic liver diseases increase the scar formation and extracellular matrix accumulation that further progress to liver fibrosis and cirrhosis. Nevertheless, there is no antifibrotic therapy to date. The ketogenic diet is composed of high fat, moderate to low-protein, and very low carbohydrate content. It is mainly used in epilepsy and Alzheimer’s disease. However, the effects of the ketogenic diet on liver fibrosis remains unknown. Through ketogenic diet consumption, β-hydroxybutyrate (bHB) and acetoacetate (AcAc) are two ketone bodies that are mainly produced in the liver. It is reported that bHB and AcAc treatment decreases cancer cell proliferation and promotes apoptosis. However, the influence of bHB and AcAc in hepatic stellate cell (HSC) activation and liver fibrosis are still unclear. Therefore, this study aimed to investigate the effect of the ketogenic diet and ketone bodies in affecting liver fibrosis progression. Our study revealed that feeding a high-fat ketogenic diet increased cholesterol accumulation in the liver, which further enhanced the carbon tetrachloride (CCl4)- and thioacetamide (TAA)-induced liver fibrosis. In addition, more severe liver inflammation and the loss of hepatic antioxidant and detoxification ability were also found in ketogenic diet-fed fibrotic mouse groups. However, the treatment with ketone bodies (bHB and AcAc) did not suppress transforming growth factor-β (TGF-β)-induced HSC activation, platelet-derived growth factor (PDGF)-BB-triggered proliferation, and the severity of CCl4-induced liver fibrosis in mice. In conclusion, our study demonstrated that feeding a high-fat ketogenic diet may trigger severe steatohepatitis and thereby promote liver fibrosis progression. Since a different ketogenic diet composition may exert different metabolic effects, more evidence is necessary to clarify the effects of a ketogenic diet on disease treatment.
Collapse
|
18
|
Rühlmann C, Dannehl D, Brodtrück M, Adams AC, Stenzel J, Lindner T, Krause BJ, Vollmar B, Kuhla A. Neuroprotective Effects of the FGF21 Analogue LY2405319. J Alzheimers Dis 2021; 80:357-369. [PMID: 33554901 DOI: 10.3233/jad-200837] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND To date, there are no effective treatments for Alzheimer's disease (AD). Thus, a significant need for research of therapies remains. OBJECTIVE One promising pharmacological target is the hormone fibroblast growth factor 21 (FGF21), which is thought to be neuroprotective. A clinical candidate for medical use could be the FGF21 analogue LY2405319 (LY), which has a specificity and potency comparable to FGF21. METHODS The present study investigated the potential neuroprotective effect of LY via PPARγ/apoE/abca1 pathway, which is known to degrade amyloid-β (Aβ) plaques by using primary glial cells and hippocampal organotypic brain slice cultures (OBSCs) from 30- and 50-week-old transgenic APPswe/PS1dE9 (tg) mice. By LY treatment of 52-week-old tg mice with advanced Aβ deposition, we further aimed to elaborate the effect of LY on AD pathology in vivo. RESULTS LY application to primary glial cells caused an upregulation of pparγ, apoE, and abca1 mRNA expression and significantly decreased number and area of Aβ plaques in OBSCs. LY treatment in tg mice increased cerebral [18F] FDG uptake and N-acetylaspartate/creatine ratio indicating enhanced neuronal activity and integrity. Although LY did not reduce the number of Aβ plaques in tg mice, the number of iba1-positive cells was significantly decreased indicating reduced microgliosis. CONCLUSION These data identified LY in vitro as an activator of Aβ degrading genes leading to cerebral Aβ load amelioration in early and late AD pathology. Although Aβ plaque reduction by LY failed in vivo, LY may be used as therapeutic agent to treat AD-related neuroinflammation and impaired neuronal integrity.
Collapse
Affiliation(s)
- Claire Rühlmann
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - David Dannehl
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Marcus Brodtrück
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Andrew C Adams
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - Bernd J Krause
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany.,Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany.,Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
19
|
Tsuchiya Y, Ozai R, Sugino T, Kawashima K, Kushibiki S, Kim YH, Sato S. Changes in peripheral blood oxidative stress markers and hepatic gene expression related to oxidative stress in Holstein cows with and without subacute ruminal acidosis during the periparturient period. J Vet Med Sci 2020; 82:1529-1536. [PMID: 32893200 PMCID: PMC7653322 DOI: 10.1292/jvms.20-0426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We investigated changes in peripheral blood metabolites, oxidative stress markers
(malondialdehyde, potential antioxidant capacity, and glutathione peroxidase [GPX]), and
hepatic gene expression related to oxidative stress in Holstein cows with and without
subacute ruminal acidosis (SARA) during the periparturient period. Eighteen multiparous
Holstein cows were categorized into SARA (n=9) or non-SARA (n=9) groups depending on
whether they developed SARA; reticulo-ruminal pH was <5.6 for more than 3 hr per day,
during the 2 weeks after parturition. Blood and liver tissue samples were collected 3
weeks prepartum and 2 and 6 weeks postpartum, with an additional blood sample collected 0
and 4 weeks postpartum. Blood aspartate transaminase (AST) and nonesterified fatty acid
(NEFA) increased significantly (P<0.05) after parturition in both
groups. GPX activity decreased gradually after parturition in the SARA group. In the SARA
group, gene expression of GPX 1 and microsomal glutathione S-transferase
3 (MGST3) decreased significantly (P<0.05), and
expression of metallothionein 2A increased significantly (P<0.05)
after parturition in the SARA group. Superoxide dismutase 1 and MGST3
decreased significantly (P<0.05) 2 weeks postpartum in the non-SARA
group. Gene expression related to oxidative stress was negatively correlated with AST,
NEFA and total ketone body levels. Therefore, the hepatic gene expression related to
oxidative stress might change associated with a negative energy balance, and might relate
the high oxidative stress in the SARA group during periparturient period.
Collapse
Affiliation(s)
- Yoshiyuki Tsuchiya
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan.,Yamagata Prefectural Agricultural Mutual Aid Association, Tendo, Yamagata 994-8511, Japan
| | - Reiko Ozai
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Toshihisa Sugino
- The Research Center for Animal Science, Graduate School of Integrated Science for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Kenji Kawashima
- Chiba Prefectural Livestock Research Center, Yachimata, Chiba 289-1113, Japan
| | - Shiro Kushibiki
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki 305-0901, Japan
| | - Yo-Han Kim
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Shigeru Sato
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan.,Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| |
Collapse
|
20
|
Kuhla A, Brichmann E, Rühlmann C, Thiele R, Meuth L, Vollmar B. Metformin Therapy Aggravates Neurodegenerative Processes in ApoE-/- Mice. J Alzheimers Dis 2020; 68:1415-1427. [PMID: 30909226 DOI: 10.3233/jad-181017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Epidemiological studies suggest that individuals with diabetes mellitus are at greater risk of developing Alzheimer's disease. A well-known insulin-sensitizing drug and the most widely prescribed oral medication for diabetes is metformin. There is evidence that metformin acts in a neuroprotective manner via the AMPK/mTOR pathway by inhibiting the tau phosphorylation. In addition, it is known that metformin upregulates Fgf21, which in turn activates the AMPK/mTOR pathway and mediates neuroprotection. Thus, metformin-induced Fgf21 release may be involved in AMPK/mTOR activation. However, some studies reported that metformin causes cognition impairment. Due to the controversial data on the neuroprotective properties of metformin, we treated Apolipoprotein E deficient (ApoE- /-) mice, a mouse model of tauopathy, with metformin for 18 weeks. Metformin-treated mice revealed increased expression of lipogenic genes, i.e., lxrα and srebp1c. In line with this, metformin caused an increase in plasma triglyceride leading to enhanced gliosis as indicated by an increase of GFAP-positive cells. Although the systemic Fgf21 concentration was increased, metformin did not activate the FgfR1c/AMPK/mTOR pathway suggesting a Fgf21-resistant state. Further, metformin-treated mice showed increased tau phosphorylation and reduced numbers of NeuN-and PSD95-positive cells. Thus, metformin-associated lipogenesis as well as inflammation aggravated neurodegenerative processes in ApoE- /- mice. Consequently, this study supports previous observations showing that metformin causes impairment of cognition.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Elaine Brichmann
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Claire Rühlmann
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Robin Thiele
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Lou Meuth
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
21
|
Hydes TJ, Ravi S, Loomba R, E Gray M. Evidence-based clinical advice for nutrition and dietary weight loss strategies for the management of NAFLD and NASH. Clin Mol Hepatol 2020; 26:383-400. [PMID: 32674529 PMCID: PMC7641567 DOI: 10.3350/cmh.2020.0067] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide and affects approximately one third of adults in the United States. The disease is becoming a global epidemic as a result of the rising rates of obesity and metabolic disease. Emerging data suggest weight loss of ≥10% overall body weight is beneficial in resolving steatosis and reversing fibrosis. Prospective trials comparing various diets are limited by lack of sufficient power as well as pre- and post-treatment histopathology, and therefore no specific diet is recommended at this time. In this narrative review we examine the pathophysiology behind specific macronutrient components that can either promote or reverse NAFLD to help inform more specific dietary recommendations. Overall, the data supports reducing saturated fat, refined carbohydrates, and red and processed meats in the diet, and increasing the consumption of plant-based foods. Diets that incorporate these recommendations include plant-based diets such as the Dietary Approaches to Stop Hypertension, Mediterranean, vegetarian, and vegan diets.
Collapse
Affiliation(s)
- Theresa J Hydes
- Division of Gastroenterology and Hepatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sujan Ravi
- Division of Gastroenterology and Hepatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rohit Loomba
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Meagan E Gray
- Division of Gastroenterology and Hepatology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
22
|
Shaito A, Hasan H, Habashy KJ, Fakih W, Abdelhady S, Ahmad F, Zibara K, Eid AH, El-Yazbi AF, Kobeissy FH. Western diet aggravates neuronal insult in post-traumatic brain injury: Proposed pathways for interplay. EBioMedicine 2020; 57:102829. [PMID: 32574954 PMCID: PMC7317220 DOI: 10.1016/j.ebiom.2020.102829] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury (TBI) is a global health burden and a major cause of disability and mortality. An early cascade of physical and structural damaging events starts immediately post-TBI. This primary injury event initiates a series of neuropathological molecular and biochemical secondary injury sequelae, that last much longer and involve disruption of cerebral metabolism, mitochondrial dysfunction, oxidative stress, neuroinflammation, and can lead to neuronal damage and death. Coupled to these events, recent studies have shown that lifestyle factors, including diet, constitute additional risk affecting TBI consequences and neuropathophysiological outcomes. There exists molecular cross-talk among the pathways involved in neuronal survival, neuroinflammation, and behavioral outcomes, that are shared among western diet (WD) intake and TBI pathophysiology. As such, poor dietary intake would be expected to exacerbate the secondary damage in TBI. Hence, the aim of this review is to discuss the pathophysiological consequences of WD that can lead to the exacerbation of TBI outcomes. We dissect the role of mitochondrial dysfunction, oxidative stress, neuroinflammation, and neuronal injury in this context. We show that currently available data conclude that intake of a diet saturated in fats, pre- or post-TBI, aggravates TBI, precludes recovery from brain trauma, and reduces the response to treatment.
Collapse
Affiliation(s)
- Abdullah Shaito
- Department of Biological and Chemical Sciences, Lebanese International University, Beirut, Lebanon and Faculty of Health Sciences, University of Balamand, Beirut, Lebanon
| | - Hiba Hasan
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | | | - Walaa Fakih
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fatima Ahmad
- Neuroscience Research Center, Faculty of Medicine, Lebanese University
| | - Kazem Zibara
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Biomedical Sciences, College of Health Sciences, Doha, Qatar
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt.
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
23
|
FGF21 Mimics a Fasting-Induced Metabolic State and Increases Appetite in Zebrafish. Sci Rep 2020; 10:6993. [PMID: 32332781 PMCID: PMC7181725 DOI: 10.1038/s41598-020-63726-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a member of the FGF superfamily that acts in an endocrine manner. FGF21 is a key regulator of energy balance and metabolism in mammals, and has emerged as a therapeutic potential for treating obesity and diabetes. Here, we report that mRNAs encoding FGF21 and its receptors are widely distributed within the zebrafish tissues and are importantly modulated by fasting (decreased in brain and liver, and increased in gut). FGF21 stimulates food intake in zebrafish, likely in part by modulating brain npy/agrp and nucb2/nesfatin-1 and gut ghrelin and cck mRNA expression. In accordance with this orexigenic role, the expression of FGF21 and its receptors were observed to increase preprandially and decrease post-feeding in the foregut and/or liver. Finally, we found important evidence in favor of a role for FGF21 in regulating glucose and lipid metabolism in the zebrafish liver in a way that mimics a fasting metabolic state.
Collapse
|
24
|
Belete TM. A Recent Achievement In the Discovery and Development of Novel Targets for the Treatment of Type-2 Diabetes Mellitus. J Exp Pharmacol 2020; 12:1-15. [PMID: 32021494 PMCID: PMC6959499 DOI: 10.2147/jep.s226113] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2DM) is a chronic metabolic disorder. Impaired insulin secretion, enhanced hepatic glucose production, and suppressed peripheral glucose use are the main defects responsible for developing the disease. Besides, the pathophysiology of T2DM also includes enhanced glucagon secretion, decreased incretin secretion, increased renal glucose reabsorption, and adipocyte, and brain insulin resistance. The increasing prevalence of T2DM in the world beseeches an urgent need for better treatment options. The antidiabetic drugs focus on control of blood glucose concentration, but the future treatment goal is to delay disease progression and treatment failure, which causes poorer glycemic regulation. Recent treatment approaches target on several novel pathophysiological defects present in T2DM. Some of the promising novel targets being under clinical development include those that increase insulin sensitization (antagonists of glucocorticoids receptor), decreasing hepatic glucose production (glucagon receptor antagonist, inhibitors of glycogen phosphorylase and fructose-1,6-biphosphatase). This review summarizes studies that are available on novel targets being studied to treat T2DM with an emphasis on the small molecule drug design. The experience gathered from earlier studies and knowledge of T2DM pathways can guide the anti-diabetic drug development toward the discovery of drugs essential to treat T2DM.
Collapse
Affiliation(s)
- Tafere Mulaw Belete
- Department of Pharmacology, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
25
|
Fibroblast growth Factor-21 promotes ketone body utilization in neurons through activation of AMP-dependent kinase. Mol Cell Neurosci 2019; 101:103415. [DOI: 10.1016/j.mcn.2019.103415] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
|
26
|
Sherrier M, Li H. The impact of keto-adaptation on exercise performance and the role of metabolic-regulating cytokines. Am J Clin Nutr 2019; 110:562-573. [PMID: 31347659 DOI: 10.1093/ajcn/nqz145] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
The ketogenic diet (KD) is a normocaloric diet composed of high-fat, low-carbohydrate, and adequate protein that induces fasting-like effects and results in the production of ketone bodies. Initially used widely for children with refractory epilepsy, the KD gained popularity due to its beneficial effects on weight loss, diabetes, and cancer. In recent years, there has been a resurgence in interest surrounding the KD and exercise performance. This review provides new insights into the adaptation period necessary for enhancement in skeletal muscle fat and ketone oxidation after sustained nutritional ketosis. In addition, this review highlights metabolically active growth factors and cytokines, which may function as important regulators of keto-adaptation in the setting of exercise and the KD.
Collapse
Affiliation(s)
- Matthew Sherrier
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Hongshuai Li
- Musculoskeletal Growth and Regeneration Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
27
|
Rose AJ. Role of Peptide Hormones in the Adaptation to Altered Dietary Protein Intake. Nutrients 2019; 11:E1990. [PMID: 31443582 PMCID: PMC6770041 DOI: 10.3390/nu11091990] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 12/25/2022] Open
Abstract
Dietary protein profoundly influences organismal traits ultimately affecting healthspan. While intracellular signalling downstream of altered amino acid supply is undoubtedly important, peptide hormones have emerged as critical factors determining systemic responses to variations in protein intake. Here the regulation and role of certain peptides hormones in such responses to altered dietary protein intake is reviewed.
Collapse
Affiliation(s)
- Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Australia.
| |
Collapse
|
28
|
Morita M, Siddiqui N, Katsumura S, Rouya C, Larsson O, Nagashima T, Hekmatnejad B, Takahashi A, Kiyonari H, Zang M, St-Arnaud R, Oike Y, Giguère V, Topisirovic I, Okada-Hatakeyama M, Yamamoto T, Sonenberg N. Hepatic posttranscriptional network comprised of CCR4-NOT deadenylase and FGF21 maintains systemic metabolic homeostasis. Proc Natl Acad Sci U S A 2019; 116:7973-7981. [PMID: 30926667 PMCID: PMC6475422 DOI: 10.1073/pnas.1816023116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Whole-body metabolic homeostasis is tightly controlled by hormone-like factors with systemic or paracrine effects that are derived from nonendocrine organs, including adipose tissue (adipokines) and liver (hepatokines). Fibroblast growth factor 21 (FGF21) is a hormone-like protein, which is emerging as a major regulator of whole-body metabolism and has therapeutic potential for treating metabolic syndrome. However, the mechanisms that control FGF21 levels are not fully understood. Herein, we demonstrate that FGF21 production in the liver is regulated via a posttranscriptional network consisting of the CCR4-NOT deadenylase complex and RNA-binding protein tristetraprolin (TTP). In response to nutrient uptake, CCR4-NOT cooperates with TTP to degrade AU-rich mRNAs that encode pivotal metabolic regulators, including FGF21. Disruption of CCR4-NOT activity in the liver, by deletion of the catalytic subunit CNOT6L, increases serum FGF21 levels, which ameliorates diet-induced metabolic disorders and enhances energy expenditure without disrupting bone homeostasis. Taken together, our study describes a hepatic CCR4-NOT/FGF21 axis as a hitherto unrecognized systemic regulator of metabolism and suggests that hepatic CCR4-NOT may serve as a target for devising therapeutic strategies in metabolic syndrome and related morbidities.
Collapse
Affiliation(s)
- Masahiro Morita
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229;
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Institute of Resource Development and Analysis, Kumamoto University, 860-0811 Kumamoto, Japan
| | - Nadeem Siddiqui
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Sakie Katsumura
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Christopher Rouya
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Ola Larsson
- Department of Oncology-Pathology, Scilifelab, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Takeshi Nagashima
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Kanagawa, Japan
| | - Bahareh Hekmatnejad
- Research Centre, Shriners Hospital for Children-Canada, Montreal, QC H4A 0A9, Canada
- Department of Human Genetics, McGill University, Montreal, QC H3A 2T5, Canada
| | - Akinori Takahashi
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, 904-0495 Okinawa, Japan
| | - Hiroshi Kiyonari
- Laboratories for Animal Resource Development and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047 Hyogo, Japan
| | - Mengwei Zang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - René St-Arnaud
- Research Centre, Shriners Hospital for Children-Canada, Montreal, QC H4A 0A9, Canada
- Department of Human Genetics, McGill University, Montreal, QC H3A 2T5, Canada
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 860-8556 Kumamoto, Japan
| | - Vincent Giguère
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H2W 1S6, Canada
| | - Ivan Topisirovic
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H2W 1S6, Canada
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Mariko Okada-Hatakeyama
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Kanagawa, Japan
- Laboratory of Cell Systems, Institute for Protein Research, Osaka University, Suita, 565-0871 Osaka, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, 904-0495 Okinawa, Japan;
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada;
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| |
Collapse
|
29
|
FGF21 Levels in Pheochromocytoma/Functional Paraganglioma. Cancers (Basel) 2019; 11:cancers11040485. [PMID: 30959789 PMCID: PMC6520899 DOI: 10.3390/cancers11040485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 01/16/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a hepatokine with beneficial effects on metabolism. Our aim was to evaluate the relationship between the serum FGF21, and energy and glucose metabolism in 40 patients with pheochromocytoma/functional paraganglioma (PPGL), in comparison with 21 obese patients and 26 lean healthy controls. 27 patients with PPGL were examined one year after tumor removal. Basic anthropometric and biochemical measurements were done. Energy metabolism was measured by indirect calorimetry (Vmax-Encore 29N). FGF21 was measured by ELISA. FGF21 was higher in PPGL than in controls (174.2 (283) pg/mL vs. 107.9 (116) pg/mL; p < 0.001) and comparable with obese (174.2 (283) pg/mL vs. 160.4 (180); p = NS). After tumor removal, FGF21 decreased (176.4 (284) pg/mL vs. 131.3 (225) pg/mL; p < 0.001). Higher levels of FGF21 were expressed, particularly in patients with diabetes. FGF21 positively correlated in PPGL with age (p = 0.005), BMI (p = 0.028), glycemia (p = 0.002), and glycated hemoglobin (p = 0.014). In conclusion, long-term catecholamine overproduction in PPGL leads to the elevation in serum FGF21, especially in patients with secondary diabetes. FGF21 levels were comparable between obese and PPGL patients, despite different anthropometric indices. We did not find a relationship between FGF21 and hypermetabolism in PPGL. Tumor removal led to the normalization of FGF21 and the other metabolic abnormalities.
Collapse
|
30
|
Nutritional Regulation of Gene Expression: Carbohydrate-, Fat- and Amino Acid-Dependent Modulation of Transcriptional Activity. Int J Mol Sci 2019; 20:ijms20061386. [PMID: 30893897 PMCID: PMC6470599 DOI: 10.3390/ijms20061386] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022] Open
Abstract
The ability to detect changes in nutrient levels and generate an adequate response to these changes is essential for the proper functioning of living organisms. Adaptation to the high degree of variability in nutrient intake requires precise control of metabolic pathways. Mammals have developed different mechanisms to detect the abundance of nutrients such as sugars, lipids and amino acids and provide an integrated response. These mechanisms include the control of gene expression (from transcription to translation). This review reports the main molecular mechanisms that connect nutrients’ levels, gene expression and metabolism in health. The manuscript is focused on sugars’ signaling through the carbohydrate-responsive element binding protein (ChREBP), the role of peroxisome proliferator-activated receptors (PPARs) in the response to fat and GCN2/activating transcription factor 4 (ATF4) and mTORC1 pathways that sense amino acid concentrations. Frequently, alterations in these pathways underlie the onset of several metabolic pathologies such as obesity, insulin resistance, type 2 diabetes, cardiovascular diseases or cancer. In this context, the complete understanding of these mechanisms may improve our knowledge of metabolic diseases and may offer new therapeutic approaches based on nutritional interventions and individual genetic makeup.
Collapse
|
31
|
Cai G, Liu J, Wang M, Su L, Cai M, Huang K, Li X, Li M, Wang L, Huang X. Mutual promotion of FGF21 and PPARγ attenuates hypoxia-induced pulmonary hypertension. Exp Biol Med (Maywood) 2019; 244:252-261. [PMID: 30714402 DOI: 10.1177/1535370219828692] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
IMPACT STATEMENT In this study, we reported for the first time that FGF21 alleviated hypoxia-induced pulmonary hypertension through attenuation of increased pulmonary arterial pressure, pulmonary arterial remodeling and collagen deposition in vivo, and we confirmed the mutual promotion of FGF21 and PPARγ in hypoxia-induced pulmonary hypertension. Additionally, we found that FGF21 and PPARγ mutually promote each other's expression via the AMPK/PGC-1α pathway and KLB protein in vitro and in vivo. Pulmonary hypertension is a progressive and serious pathological phenomenon with a poor prognosis, and current therapies are highly limited. Our results provide novel insight into potential clinical therapies for pulmonary hypertension and establish the possibility of using this drug combination and potential dosage reductions in clinical settings.
Collapse
Affiliation(s)
- Gexiang Cai
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Jingjing Liu
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Meibin Wang
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Lihuang Su
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Mengsi Cai
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Kate Huang
- 2 Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang 325000, PR China
| | - Xiuchun Li
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Manxiang Li
- 3 Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Shanxi 710061, PR China
| | - Liangxing Wang
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| | - Xiaoying Huang
- 1 Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, PR China
| |
Collapse
|
32
|
Zilberter T, Zilberter Y. Ketogenic Ratio Determines Metabolic Effects of Macronutrients and Prevents Interpretive Bias. Front Nutr 2018; 5:75. [PMID: 30214902 PMCID: PMC6125377 DOI: 10.3389/fnut.2018.00075] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022] Open
|
33
|
Zhang Y, Higgins CB, Mayer AL, Mysorekar IU, Razani B, Graham MJ, Hruz PW, DeBosch BJ. TFEB-dependent induction of thermogenesis by the hepatocyte SLC2A inhibitor trehalose. Autophagy 2018; 14:1959-1975. [PMID: 29996716 PMCID: PMC6152536 DOI: 10.1080/15548627.2018.1493044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 06/13/2018] [Accepted: 06/18/2018] [Indexed: 02/08/2023] Open
Abstract
The macroautophagy/autophagy-inducing disaccharide, trehalose, has been proposed to be a promising therapeutic agent against neurodegenerative and cardiometabolic diseases. We recently showed that trehalose attenuates hepatic steatosis in part by blocking hepatocyte glucose transport to induce hepatocyte autophagic flux. However, although every major demonstration of trehalose action invokes activating autophagic flux as its primary function, the mechanism of action of trehalose in whole-body energy metabolism remains poorly defined. Here, we demonstrate that trehalose induces hepatocyte TFEB (transcription factor EB)-dependent thermogenesis in vivo, concomitant with upregulation of hepatic and white adipose expression of UCP1 (uncoupling protein 1 [mitochondrial, protein carrier]). Mechanistically, we provide evidence that hepatocyte fasting transcriptional and metabolic responses depend upon PPARGC1A (peroxisome proliferative activated receptor, gamma, coactivator 1 alpha), TFEB, and FGF21 (fibroblast growth factor 21) signaling. Strikingly, hepatocyte-selective TFEB knockdown abrogated trehalose induction of thermogenesis and white adipose tissue UCP1 upregulation in vivo. In contrast, we found that trehalose action on thermogenesis was independent of LEP (leptin) and the autophagy pathway, as there was robust thermogenic induction in trehalose-treated ob/ob, Becn1, Atg16l1, and Epg5 mutant mice. We conclude that trehalose induces metabolically favorable effects on whole-body thermogenesis in part via hepatocyte-centered fasting-like mechanisms that appear to be independent of autophagic flux. Our findings elucidate a novel mechanism by which trehalose acts as a metabolic therapeutic agent by activating hepatic fasting responses. More broadly, the hepatic glucose fasting response may be of clinical utility against overnutrition-driven disease, such as obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Yiming Zhang
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Allyson L. Mayer
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Indira U. Mysorekar
- Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Babak Razani
- Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark J. Graham
- IONIS Pharmaceuticals, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul W. Hruz
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian J. DeBosch
- Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Obesity is a global health crisis with detrimental effects on all organ systems leading to worsening disease state and rising costs of care. Persons with obesity failing lifestyle therapies need to be escalated to appropriate pharmacological treatment modalities, medical devices, and/or bariatric surgery if criteria are met and more aggressive intervention is needed. The progression of severe obesity in the patient population coupled with related co-morbidities necessitates the development of novel therapies for the treatment of obesity. This development is preceded by increased understanding of the underpinnings of energy regulation and neurohormonal pathways involved in energy homeostasis. RECENT FINDINGS Though there are approved anti-obesity drugs available in the USA, newer drugs are now in the pipeline for development given the urgent need. This review focuses on anti-obesity drugs in the pipeline including centrally acting agents (setmelanotide, neuropeptide Y antagonist [velneperit], zonisamide-bupropion [Empatic], cannabinoid type-1 receptor blockers), gut hormones and incretin targets (new glucagon-like-peptide-1 [GLP-1] analogues [semaglutide and oral equivalents], amylin mimetics [davalintide, dual amylin and calcitonin receptor agonists], dual action GLP-1/glucagon receptor agonists [oxyntomodulin], triple agonists [tri-agonist 1706], peptide YY, leptin analogues [combination pramlintide-metreleptin]), and other novel targets (methionine aminopeptidase 2 inhibitor [beloranib], lipase inhibitor [cetilistat], triple monoamine reuptake inhibitor [tesofensine], fibroblast growth factor 21), including anti-obesity vaccines (ghrelin, somatostatin, adenovirus36). With these new drugs in development, anti-obesity therapeutics have potential to vastly expand allowing better treatment options and personalized approach to obesity care.
Collapse
Affiliation(s)
- Gitanjali Srivastava
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine, 720 Harrison Avenue, 8th Floor, Suite 801, Boston, MA, 02118, USA
| | - Caroline Apovian
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine, 720 Harrison Avenue, 8th Floor, Suite 801, Boston, MA, 02118, USA.
| |
Collapse
|
35
|
Ryan KK, Packard AEB, Larson KR, Stout J, Fourman SM, Thompson AMK, Ludwick K, Habegger KM, Stemmer K, Itoh N, Perez-Tilve D, Tschöp MH, Seeley RJ, Ulrich-Lai YM. Dietary Manipulations That Induce Ketosis Activate the HPA Axis in Male Rats and Mice: A Potential Role for Fibroblast Growth Factor-21. Endocrinology 2018; 159:400-413. [PMID: 29077838 PMCID: PMC5761593 DOI: 10.1210/en.2017-00486] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/20/2017] [Indexed: 12/19/2022]
Abstract
In response to an acute threat to homeostasis or well-being, the hypothalamic-pituitary-adrenocortical (HPA) axis is engaged. A major outcome of this HPA axis activation is the mobilization of stored energy, to fuel an appropriate behavioral and/or physiological response to the perceived threat. Importantly, the extent of HPA axis activity is thought to be modulated by an individual's nutritional environment. In this study, we report that nutritional manipulations signaling a relative depletion of dietary carbohydrates, thereby inducing nutritional ketosis, acutely and chronically activate the HPA axis. Male rats and mice maintained on a low-carbohydrate high-fat ketogenic diet (KD) exhibited canonical markers of chronic stress, including increased basal and stress-evoked plasma corticosterone, increased adrenal sensitivity to adrenocorticotropin hormone, increased stress-evoked c-Fos immunolabeling in the paraventricular nucleus of the hypothalamus, and thymic atrophy, an indicator of chronic glucocorticoid exposure. Moreover, acutely feeding medium-chain triglycerides (MCTs) to rapidly induce ketosis among chow-fed male rats and mice also acutely increased HPA axis activity. Lastly, and consistent with a growing literature that characterizes the hepatokine fibroblast growth factor-21 (FGF21) as both a marker of the ketotic state and as a key metabolic stress hormone, the HPA response to both KD and MCTs was significantly blunted among mice lacking FGF21. We conclude that dietary manipulations that induce ketosis lead to increased HPA axis tone, and that the hepatokine FGF21 may play an important role to facilitate this effect.
Collapse
Affiliation(s)
- Karen K. Ryan
- Department of Neurobiology, Physiology & Behavior, University of California, Davis, California 95616
| | - Amy E. B. Packard
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio 45237
| | - Karlton R. Larson
- Department of Neurobiology, Physiology & Behavior, University of California, Davis, California 95616
| | - Jayna Stout
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio 45237
| | - Sarah M. Fourman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio 45237
| | - Abigail M. K. Thompson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio 45237
| | - Kristen Ludwick
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio 45237
| | - Kirk M. Habegger
- Department of Medicine, University of Alabama, Birmingham, Alabama 35294
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich & Division of Metabolic Diseases, Technische Universität München, D-85748 Munich, Germany
| | - Nobuyuki Itoh
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto 606-8501, Japan
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio 45237
| | - Matthias H. Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich & Division of Metabolic Diseases, Technische Universität München, D-85748 Munich, Germany
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Yvonne M. Ulrich-Lai
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio 45237
| |
Collapse
|
36
|
Rühlmann C, Wölk T, Blümel T, Stahn L, Vollmar B, Kuhla A. Long-term caloric restriction in ApoE-deficient mice results in neuroprotection via Fgf21-induced AMPK/mTOR pathway. Aging (Albany NY) 2017; 8:2777-2789. [PMID: 27902456 PMCID: PMC5191869 DOI: 10.18632/aging.101086] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023]
Abstract
Caloric restriction (CR) decelerates the aging process, extends lifespan and exerts neuroprotective effects in diverse species by so far unknown mechanisms. Based on known neuroprotective effects of fibroblastic growth factor 21 (Fgf21) we speculate that CR upregulates Fgf21, which phosphorylates neuronal AMP-activated protein kinase (AMPK), leading to a decrease of mammalian target of rapamycin (mTOR) signaling activity and an inhibition of tau-hyperphosphorylation. This in turn reduces the formation of neurofibrillary tangles, a neuropathological hallmark of Alzheimer's disease. ApoE-deficient mice (ApoE−/−), serving as a model of neurodegeneration, showed upon CR vs. ad libitum feeding increased Fgf21 levels in both, plasma and brain as well as higher phosphorylation of fibroblastic growth factor receptor 1c (Fgfr1c), extracellular signal-regulated kinases 1/2 (ERK1/2) and AMPK in brain, lower activity of mTOR and decreased Tau-phosphorylation. Finally, CR in ApoE−/− mice caused neuroprotection as indicated by a higher synaptic plasticity shown by immunohistochemical analysis with increased numbers of PSD95-positive neurons and a better cognitive performance as analyzed with Morris water maze test. These data provide substantial evidence that neuroprotection upon CR seems to be Fgf21-dependent. Further experiments are necessary to evaluate Fgf21 as a therapeutic tool to treat tauopathy for improvement of cognitive performance.
Collapse
Affiliation(s)
- Claire Rühlmann
- Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Tjark Wölk
- Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Tobias Blümel
- Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Laura Stahn
- Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
37
|
Gómez-Sámano MÁ, Grajales-Gómez M, Zuarth-Vázquez JM, Navarro-Flores MF, Martínez-Saavedra M, Juárez-León ÓA, Morales-García MG, Enríquez-Estrada VM, Gómez-Pérez FJ, Cuevas-Ramos D. Fibroblast growth factor 21 and its novel association with oxidative stress. Redox Biol 2017; 11:335-341. [PMID: 28039838 PMCID: PMC5200873 DOI: 10.1016/j.redox.2016.12.024] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 02/07/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is an endocrine-member of the FGF family. It is synthesized mainly in the liver, but it is also expressed in adipose tissue, skeletal muscle, and many other organs. It has a key role in glucose and lipid metabolism, as well as in energy balance. FGF21 concentration in plasma is increased in patients with obesity, insulin resistance, and metabolic syndrome. Recent findings suggest that such increment protects tissue from an increased oxidative stress environment. Different types of physical stress, such as strenuous exercising, lactation, diabetic nephropathy, cardiovascular disease, and critical illnesses, also increase FGF21 circulating concentration. FGF21 is now considered a stress-responsive hormone in humans. The discovery of an essential response element in the FGF21 gene, for the activating transcription factor 4 (ATF4), involved in the regulation of oxidative stress, and its relation with genes such as NRF2, TBP-2, UCP3, SOD2, ERK, and p38, places FGF21 as a key regulator of the oxidative stress cell response. Its role in chronic diseases and its involvement in the treatment and follow-up of these diseases has been recently the target of new studies. The diminished oxidative stress through FGF21 pathways observed with anti-diabetic therapy is another clue of the new insights of this hormone.
Collapse
Affiliation(s)
- Miguel Ángel Gómez-Sámano
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Mariana Grajales-Gómez
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Julia María Zuarth-Vázquez
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Ma Fernanda Navarro-Flores
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Mayela Martínez-Saavedra
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Óscar Alfredo Juárez-León
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Mariana G Morales-García
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Víctor Manuel Enríquez-Estrada
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Francisco J Gómez-Pérez
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Daniel Cuevas-Ramos
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
38
|
Kokkinos J, Tang S, Rye KA, Ong KL. The role of fibroblast growth factor 21 in atherosclerosis. Atherosclerosis 2017; 257:259-265. [DOI: 10.1016/j.atherosclerosis.2016.11.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/07/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
|
39
|
McCarty MF. Practical prospects for boosting hepatic production of the "pro-longevity" hormone FGF21. Horm Mol Biol Clin Investig 2015; 30:/j/hmbci.ahead-of-print/hmbci-2015-0057/hmbci-2015-0057.xml. [PMID: 26741352 DOI: 10.1515/hmbci-2015-0057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 11/20/2015] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor-21 (FGF21), produced mainly in hepatocytes and adipocytes, promotes leanness, insulin sensitivity, and vascular health while down-regulating hepatic IGF-I production. Transgenic mice overexpressing FGF21 enjoy a marked increase in median and maximal longevity comparable to that evoked by calorie restriction - but without a reduction in food intake. Transcriptional factors which promote hepatic FGF21 expression include PPARα, ATF4, STAT5, and FXR; hence, fibrate drugs, elevated lipolysis, moderate-protein vegan diets, growth hormone, and bile acids may have potential to increase FGF21 synthesis. Sirt1 activity is required for optimal responsiveness of FGF21 to PPARα, and Sirt1 activators can boost FGF21 transcription. Conversely, histone deacetylase 3 (HDAC3) inhibits PPARα's transcriptional impact on FGF21, and type 1 deacetylase inhibitors such as butyrate therefore increase FGF21 expression. Glucagon-like peptide-1 (GLP-1) increases hepatic expression of both PPARα and Sirt1; acarbose, which increases intestinal GLP-1 secretion, also increases FGF21 and lifespan in mice. Glucagon stimulates hepatic production of FGF21 by increasing the expression of the Nur77 transcription factor; increased glucagon secretion can be evoked by supplemental glycine administered during post-absorptive metabolism. The aryl hydrocarbon receptor (AhR) has also been reported recently to promote FGF21 transcription. Bilirubin is known to be an agonist for this receptor, and this may rationalize a recent report that heme oxygenase-1 induction in the liver boosts FGF21 expression. There is reason to suspect that phycocyanorubin, a bilirubin homolog that is a metabolite of the major phycobilin in spirulina, may share bilirubin's agonist activity for AhR, and perhaps likewise promote FGF21 induction. In the future, regimens featuring a plant-based diet, nutraceuticals, and safe drugs may make it feasible to achieve physiologically significant increases in FGF21 that promote metabolic health, leanness, and longevity.
Collapse
|
40
|
Akbar H, Batistel F, Drackley JK, Loor JJ. Alterations in Hepatic FGF21, Co-Regulated Genes, and Upstream Metabolic Genes in Response to Nutrition, Ketosis and Inflammation in Peripartal Holstein Cows. PLoS One 2015; 10:e0139963. [PMID: 26451842 PMCID: PMC4599736 DOI: 10.1371/journal.pone.0139963] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/18/2015] [Indexed: 12/29/2022] Open
Abstract
In rodents, fibroblast growth factor 21 (FGF21) has emerged as a key metabolic regulator produced by liver. To gather preliminary data on the potential importance of FGF1, co-regulated genes, and upstream metabolic genes, we examined the hepatic mRNA expression in response to nutrition and inflammation in dairy cows. In experiment 1, induction of ketosis through feed restriction on d 5 postpartum upregulated FGF21, its co-receptor KLB, and PPARA but only elicited a numerical increase in serum FGF21 concentration. In experiment 2, cows in control (CON) or receiving 50 g/d of L-carnitine (C50) from -14 through 21 d had increased FGF21, PPARA, and NFIL3 on d 10 compared with d 2 postpartum. In contrast, compared with CON and C50, 100 g/d L-carnitine (C100) resulted in lower FGF21, KLB, ANGPTL4, and ARNTL expression on d 10. In experiment 3, cows were fed during the dry period either a higher-energy (OVE; 1.62 Mcal/kg DM) or lower-energy (CON; 1.34 Mcal/kg DM) diet and received 0 (OVE:N, CON:N) or 200 μg of LPS (OVE:Y, CON:Y) into the mammary gland at d 7 postpartum. For FGF21 mRNA expression in CON, the LPS challenge (CON:Y) prevented a decrease in expression between d 7 and 14 postpartum such that cows in CON:N had a 4-fold lower expression on d 14 compared with d 7. The inflammatory stimulus induced by LPS in CON:Y resulted in upregulation of PPARA on d 14 to a similar level as cows in OVE:N. In OVE:Y, expression of PPARA was lower than CON:N on d 7 and remained unchanged on d 14. On d 7, LPS led to a 4-fold greater serum FGF21 only in OVE but not in CON cows. In fact, OVE:Y reached the same serum FGF21 concentration as CON:N, suggesting a carryover effect of dietary energy level on signaling mechanisms within liver. Overall, results indicate that nutrition, ketosis, and inflammation during the peripartal period can alter hepatic FGF21, co-regulated genes, and upstream metabolic genes to various extents. The functional outcome of these changes merits further study, and in particular the mechanisms regulating transcription in response to changes in energy balance and feed intake.
Collapse
Affiliation(s)
- Haji Akbar
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Fernanda Batistel
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - James K. Drackley
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Juan J. Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
Suboptimal maternal nutrition exerts lasting impacts on obesity risk in offspring, but the direction of the effect is determined by the timing of exposure. While maternal undernutrition in early pregnancy is associated with increased body mass index, in later pregnancy it can be protective. The importance of the timing of maternal undernutrition is also observed in rodents, however, many of the processes that occur in the last trimester of human gestation are delayed to the postnatal period. Neonatal leptin administration exerts lasting impacts on susceptibility to obesity in rodents. Although leptin can influence the formation of hypothalamic circuits involved in homeostatic control of feeding during the postnatal period, these effects are too late to account for its ability to reverse adverse metabolic programming due to early gestational exposure to maternal undernutrition. This review presents an alternative framework for understanding the effects of neonatal leptin through influences on developing thermoregulatory circuits.
Collapse
|
42
|
Domouzoglou EM, Naka KK, Vlahos AP, Papafaklis MI, Michalis LK, Tsatsoulis A, Maratos-Flier E. Fibroblast growth factors in cardiovascular disease: The emerging role of FGF21. Am J Physiol Heart Circ Physiol 2015; 309:H1029-38. [PMID: 26232236 PMCID: PMC4747916 DOI: 10.1152/ajpheart.00527.2015] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 07/22/2015] [Indexed: 01/07/2023]
Abstract
Early detection of risk factors for enhanced primary prevention and novel therapies for treating the chronic consequences of cardiovascular disease are of the utmost importance for reducing morbidity. Recently, fibroblast growth factors (FGFs) have been intensively studied as potential new molecules in the prevention and treatment of cardiovascular disease mainly attributable to metabolic effects and angiogenic actions. Members of the endocrine FGF family have been shown to increase metabolic rate, decrease adiposity, and restore glucose homeostasis, suggesting a multiple metabolic role. Serum levels of FGFs have been associated with established cardiovascular risk factors as well as with the severity and extent of coronary artery disease and could be useful for prediction of cardiovascular death. Furthermore, preclinical investigations and clinical trials have tested FGF administration for therapeutic angiogenesis in ischemic vascular disease, demonstrating a potential role in improving angina and limb function. FGF21 has lately emerged as a potent metabolic regulator with multiple effects that ultimately improve the lipoprotein profile. Early studies show that FGF21 is associated with the presence of atherosclerosis and may play a protective role against plaque formation by improving endothelial function. The present review highlights recent investigations suggesting that FGFs, in particular FGF21, may be useful as markers of cardiovascular risk and may also serve as protective/therapeutic agents in cardiovascular disease.
Collapse
Affiliation(s)
- Eleni M Domouzoglou
- Department of Pediatrics, Medical School, University of Ioannina, Ioannina, Greece
| | - Katerina K Naka
- Second Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Antonios P Vlahos
- Department of Pediatrics, Medical School, University of Ioannina, Ioannina, Greece
| | - Michail I Papafaklis
- Second Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Lampros K Michalis
- Second Department of Cardiology, Medical School, University of Ioannina, Ioannina, Greece
| | - Agathoklis Tsatsoulis
- Department of Endocrinology, Medical School, University of Ioannina, Ioannina, Greece
| | - Eleftheria Maratos-Flier
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
43
|
Mericq V, De Luca F, Hernandez MI, Peña V, Rossel K, Garcia M, Avila A, Cavada G, Iñiguez G. Serum fibroblast growth factor 21 levels are inversely associated with growth rates in infancy. Horm Res Paediatr 2015; 82:324-31. [PMID: 25300595 DOI: 10.1159/000367922] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) is a metabolic and growth regulator. AIM To investigate the role of FGF21 during growth in infancy. METHODS Blood samples for FGF21, leptin, insulin and glucose were collected from cord blood obtained from 95 preterm and term newborns (cross-sectional group), and at 6 and 12 months of life in 80 preterm and term infants (longitudinal group). Length and weight were measured at birth, 6 months, and 12 months. RESULTS From birth through 12 months of age, preterm infants' linear growth and weight gain were larger than those of term infants, irrespective of birth weight SDS. At birth and at 12 months, there was no difference in FGF21 levels between preterm and term infants; in contrast, at 6 months, serum FGF21 in term infants was significantly higher than that of preterm ones. In the 0-6-month period, in the whole longitudinal group, serum FGF21 was inversely correlated to the length change SDS, and such a significant inverse correlation persisted in the preterm-AGA group in the 6-12-month period. In addition, term infants who experienced length catch-up in the first 6 months of life exhibited lower serum FGF21 levels at 6 months, and those with length catch-up growth between 6 and 12 months had a greater decrease of serum FGF21 level in the same time period. CONCLUSIONS Our results indicate that FGF21 in infancy is inversely correlated with linear growth rate, thus suggesting that FGF21 is a negative regulator of human growth.
Collapse
Affiliation(s)
- Verónica Mericq
- Institute of Maternal and Child Research, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Zhang A, Sieglaff DH, York JP, Suh JH, Ayers SD, Winnier GE, Kharitonenkov A, Pin C, Zhang P, Webb P, Xia X. Thyroid hormone receptor regulates most genes independently of fibroblast growth factor 21 in liver. J Endocrinol 2015; 224:289-301. [PMID: 25501997 DOI: 10.1530/joe-14-0440] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Thyroid hormone (TH) acts through specific receptors (TRs), which are conditional transcription factors, to induce fibroblast growth factor 21 (FGF21), a peptide hormone that is usually induced by fasting and that influences lipid and carbohydrate metabolism via local hepatic and systemic endocrine effects. While TH and FGF21 display overlapping actions when administered, including reductions in serum lipids, according to the current models these hormones act independently in vivo. In this study, we examined mechanisms of regulation of FGF21 expression by TH and tested the possibility that FGF21 is required for induction of hepatic TH-responsive genes. We confirm that active TH (triiodothyronine (T3)) and the TRβ-selective thyromimetic GC1 increase FGF21 transcript and peptide levels in mouse liver and that this effect requires TRβ. T3 also induces FGF21 in cultured hepatocytes and this effect involves direct actions of TRβ1, which binds a TRE within intron 2 of FGF21. Gene expression profiles of WT and Fgf21-knockout mice are very similar, indicating that FGF21 is dispensable for the majority of hepatic T3 gene responses. A small subset of genes displays diminished T3 response in the absence of FGF21. However, most of these are not obviously directly involved in T3-dependent hepatic metabolic processes. Consistent with these results, T3-dependent effects on serum cholesterol are maintained in the Fgf21(-/-) background and we observe no effect of the Fgf21-knockout background on serum triglycerides and glucose. Our findings indicate that T3 regulates the genes involved in classical hepatic metabolic responses independently of FGF21.
Collapse
Affiliation(s)
- Aijun Zhang
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Douglas H Sieglaff
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Jean Philippe York
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Ji Ho Suh
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Stephen D Ayers
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Glenn E Winnier
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Alexei Kharitonenkov
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Christopher Pin
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Pumin Zhang
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Paul Webb
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Xuefeng Xia
- Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China Houston Methodist Research InstituteGenomic Medicine Program, 6670 Bertner Ave, Houston, Texas 77030, USACollege of Arts and SciencesChemistry Department, Indiana University Bloomington, Bloomington, Indiana, USADepartments of PaediatricsOncology, and Physiology and Pharmacology, University of Western Ontario, London, Ontario, CanadaChildren's Health Research InstituteLondon, Ontario, CanadaDepartment of Molecular Physiology and BiophysicsBaylor College of Medicine, Houston, Texas, USAThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| |
Collapse
|
46
|
Akbar H, Grala T, Vailati Riboni M, Cardoso F, Verkerk G, McGowan J, Macdonald K, Webster J, Schutz K, Meier S, Matthews L, Roche J, Loor J. Body condition score at calving affects systemic and hepatic transcriptome indicators of inflammation and nutrient metabolism in grazing dairy cows. J Dairy Sci 2015; 98:1019-32. [DOI: 10.3168/jds.2014-8584] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 11/03/2014] [Indexed: 12/15/2022]
|
47
|
Abstract
The ever growing prevalence of childhood obesity is being accompanied by an increase in the pediatric population of diseases once believed to be exclusive of the adulthood such as the metabolic syndrome (MS). The MS has been defined as the link between insulin resistance, hypertension, dyslipidemia, impaired glucose tolerance, and other metabolic abnormalities associated with an increased risk of atherosclerotic cardiovascular diseases in adults. In this review, we will discuss the peculiar aspects of the pediatric MS and the role of novel molecules and biomarkers in its pathogenesis.
Collapse
|
48
|
GCN2 and FGF21 are likely mediators of the protection from cancer, autoimmunity, obesity, and diabetes afforded by vegan diets. Med Hypotheses 2014; 83:365-71. [DOI: 10.1016/j.mehy.2014.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/14/2014] [Indexed: 12/20/2022]
|
49
|
|
50
|
Cyphert HA, Alonge KM, Ippagunta SM, Hillgartner FB. Glucagon stimulates hepatic FGF21 secretion through a PKA- and EPAC-dependent posttranscriptional mechanism. PLoS One 2014; 9:e94996. [PMID: 24733293 PMCID: PMC3986400 DOI: 10.1371/journal.pone.0094996] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/21/2014] [Indexed: 12/14/2022] Open
Abstract
Previous studies have shown that whole body deletion of the glucagon receptor suppresses the ability of starvation to increase hepatic fibroblast growth factor 21 (FGF21) expression and plasma FGF21 concentration. Here, we investigate the mechanism by which glucagon receptor activation increases hepatic FGF21 production. Incubating primary rat hepatocyte cultures with glucagon, dibutyryl cAMP or forskolin stimulated a 3-4-fold increase in FGF21 secretion. The effect of these agents on FGF21 secretion was not associated with an increase in FGF21 mRNA abundance. Glucagon induction of FGF21 secretion was additive with the stimulatory effect of a PPARα activator (GW7647) on FGF21 secretion. Inhibition of protein kinase A (PKA) and downstream components of the PKA pathway [i.e. AMP-activated protein kinase and p38 MAPK] suppressed glucagon activation of FGF21 secretion. Incubating hepatocytes with an exchange protein directly activated by cAMP (EPAC)-selective cAMP analog [i.e. 8-(4-chlorophenylthio)-2'-O-methyladenosine-3', 5'-cyclic monophosphate (cpTOME)], stimulated a 3.9-fold increase FGF21 secretion, whereas inhibition of the EPAC effector, Rap1, suppressed glucagon activation of FGF21 secretion. Treatment of hepatocytes with insulin also increased FGF21 secretion. In contrast to glucagon, insulin activation of FGF21 secretion was associated with an increase in FGF21 mRNA abundance. Glucagon synergistically interacted with insulin to stimulate a further increase in FGF21 secretion and FGF21 mRNA abundance. These results demonstrate that glucagon increases hepatic FGF21 secretion via a posttranscriptional mechanism and provide evidence that both the PKA branch and EPAC branch of the cAMP pathway play a role in mediating this effect. These results also identify a novel synergistic interaction between glucagon and insulin in the regulation of FGF21 secretion and FGF21 mRNA abundance. We propose that this insulin/glucagon synergism plays a role in mediating the elevation in FGF21 production during starvation and conditions related to metabolic syndrome.
Collapse
Affiliation(s)
- Holly A Cyphert
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia, United States of America
| | - Kimberly M Alonge
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia, United States of America
| | - Siri M Ippagunta
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia, United States of America
| | - F Bradley Hillgartner
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|