1
|
Kang J, Zhu JQ, Wang Y, He Q. Effect of Immunosuppressive Regimens on Metabolic Dysfunction-associated Fatty Liver Disease Following Liver Transplantation. J Clin Exp Hepatol 2025; 15:102387. [PMID: 39268481 PMCID: PMC11388780 DOI: 10.1016/j.jceh.2024.102387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/31/2024] [Indexed: 09/15/2024] Open
Abstract
Background Metabolic dysfunction-associated fatty liver disease has been linked to negative outcomes in patients with end-stage liver disease following liver transplantation. However, the influence of immunosuppressive regimens on it has not been explored. Methods A retrospective analysis was conducted using the preoperative and postoperative data from patients with end-stage liver disease. The study compared three different groups: tacrolimus-based group, sirolimus-based group, and combined tacrolimus- and sirolimus-based regimens. Binary logistic regression analysis was employed to identify risk factors for metabolic dysfunction-associated fatty liver disease. Results A total of 171 patients participated in the study, consisting of 127 males and 44 females, with a mean age of 49.6 years. The prevalence of posttransplant metabolic dysfunction-associated fatty liver disease was 29.23%. Among the three groups, there were 111 liver transplant recipients in the tacrolimus-based group, 28 in the sirolimus-based group, and 32 in the combination group. A statistically significant difference was observed in the incidence of metabolic dysfunction-associated fatty liver disease (P < 0.05), whereas the other preoperative and postoperative parameters showed no significant differences. Multivariate analysis revealed that a low-calorie diet (95% confidence intervals: 0.15-0.90, P = 0.021) and a combination of tacrolimus- and sirolimus-based immunosuppressive regimen (95% confidence intervals: 1.01-2.77, P = 0.046) were associated with lower risk of posttransplant metabolic dysfunction-associated fatty liver disease. Conclusions Our study indicates that implementing a low-calorie diet and utilizing a combination of tacrolimus- and sirolimus-based immunosuppressive regimen can effectively lower the risk of posttransplant metabolic dysfunction-associated fatty liver disease following liver transplantation.
Collapse
Affiliation(s)
- Jing Kang
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Ji-Qiao Zhu
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Yan Wang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Qiang He
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| |
Collapse
|
2
|
Guo D, Sun J, Feng S. Comparative analysis of the effects of high-intensity interval training and traditional aerobic training on improving physical fitness and biochemical indicators in patients with non-alcoholic fatty liver disease. J Sports Med Phys Fitness 2025; 65:132-139. [PMID: 39287582 DOI: 10.23736/s0022-4707.24.16206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), linked to sedentary lifestyles and metabolic dysfunction, is highly prevalent. Exercise is an established intervention, but the relative efficacy of different exercise modalities remains unclear. The aim of this study was to compare the effects of moderate-intensity continuous aerobic training and High-Intensity Interval Training (HIIT) on physical fitness, biochemical parameters, and liver function in NAFLD patients. METHODS Sixty NAFLD patients (32 males, 28 females; age: 49.7±8.7 years; BMI: 31.1±3.3 kg/m2) were randomized into HIIT, aerobic training, and control cohorts. The HIIT cohort performed 4-minute high-intensity intervals at 85-95% of peak heart rate, interspersed with 3-minute active recovery at 60-70% of peak heart rate for 30-40 minutes per session. The aerobic training cohort performed continuous exercise at 60-70% of peak heart rate for 45-60 minutes per session. Both intervention cohorts underwent 12 weeks of supervised training, thrice weekly. Before and after the intervention, assessments included cardiorespiratory fitness, muscular strength, flexibility, lipid profile, liver enzymes, inflammatory markers, insulin sensitivity, and oxidative stress markers. RESULTS Compared to controls, both exercise cohorts showed significant improvements in cardiorespiratory fitness, muscular strength, and flexibility. However, HIIT elicited superior enhancements in cardiorespiratory fitness and muscular strength. Biochemically, both exercise cohorts exhibited reductions in triglycerides, low-density lipoprotein (LDL) cholesterol, liver enzymes (alanine aminotransferase [ALT], aspartate aminotransferase [AST]), inflammatory markers (C-reactive protein [CRP], interleukin-6 [IL-6]), insulin resistance (homeostatic model assessment of insulin resistance [HOMA-IR]), and oxidative stress markers (malondialdehyde [MDA], protein carbonyl). Notably, HIIT yielded more substantial improvements in these parameters. CONCLUSIONS HIIT and traditional aerobic training are effective in improving physical fitness and ameliorating biochemical indicators in NAFLD patients. Notably, HIIT appears to be more advantageous in enhancing cardiorespiratory fitness, muscular strength, and metabolic, inflammatory, and oxidative stress profiles, suggesting its potential as a time-efficient and effective exercise modality for managing NAFLD.
Collapse
Affiliation(s)
- Dawei Guo
- Department of Physical, Shaanxi Xueqian Normal University, Xi'an, Shaanxi, China
| | - Jian Sun
- School of Martial Arts, Northeast China Ethnic Traditional Sports Research Center, Wuhan Sports University, Wuhan, China
| | - Shuolei Feng
- Lincoln University College (LUC), Petaling Jaya, Malaysia -
- College of Arts, Sciences and Education, St. Dominic Savio College, Caloocan, Philippines
| |
Collapse
|
3
|
Fodor Duric L, Belčić V, Oberiter Korbar A, Ćurković S, Vujicic B, Gulin T, Muslim J, Gulin M, Grgurević M, Catic Cuti E. The Role of SHBG as a Marker in Male Patients with Metabolic-Associated Fatty Liver Disease: Insights into Metabolic and Hormonal Status. J Clin Med 2024; 13:7717. [PMID: 39768643 PMCID: PMC11677371 DOI: 10.3390/jcm13247717] [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: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Metabolic-associated fatty liver disease (MAFLD) is a spectrum of liver diseases linked to insulin resistance (IR), type 2 diabetes, and metabolic disorders. IR accelerates fat accumulation in the liver, worsening MAFLD. Regular physical activity and weight loss can improve liver function, reduce fat, and lower cardiovascular risk. This study examines the role of sex hormone-binding globulin (SHBG) in MAFLD, focusing on its potential as a biomarker and its relationship with insulin resistance. Methods: The study included 98 male patients (ages 30-55) with MAFLD, identified through systematic examinations, and 74 healthy male controls. All participants underwent abdominal ultrasound and blood tests after fasting, assessing markers such as glucose, liver enzymes (AST, ALT, γGT), lipids (cholesterol, triglycerides), insulin, SHBG, estradiol, and testosterone. SHBG levels were analyzed in relation to body mass index (BMI) and age. Results: A significant association was found between low SHBG levels and the presence of fatty liver. Individuals with MAFLD had lower SHBG levels compared to controls. BMI and age were key factors influencing SHBG, with higher BMI linked to lower SHBG in younger men, while SHBG remained stable in older individuals regardless of BMI. Conclusion: SHBG may serve as a valuable biomarker for early detection and risk assessment of MAFLD. The complex relationship between SHBG, BMI, and age highlights the importance of considering both hormonal and metabolic factors when assessing fatty liver risk. Our findings support the need for comprehensive metabolic evaluations in clinical practice.
Collapse
Affiliation(s)
- Ljiljana Fodor Duric
- School of Medicine, University of Catholica Croatica, 10000 Zagreb, Croatia
- Department of Nephrology and Arterial Hypertension, Medikol Polyclinic, 10000 Zagreb, Croatia
| | - Velimir Belčić
- Medikol Polyclinic, 10000 Zagreb, Croatia; (V.B.); (J.M.)
| | | | - Sanja Ćurković
- Faculty of Kinesiology, University of Zagreb, 10000 Zagreb, Croatia;
- Faculty of Agriculture, University of Zagreb, 10000 Zagreb, Croatia
| | - Bozidar Vujicic
- School of Medicine, University of Rijeka, 10000 Rijeka, Croatia;
- Department of Nephrology, Dialysis and Transplantation, University Hospital Center Rijeka, 10000 Rijeka, Croatia
| | - Tonko Gulin
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
- Department of Nephrology and Arterial Hypertension, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
| | - Jelena Muslim
- Medikol Polyclinic, 10000 Zagreb, Croatia; (V.B.); (J.M.)
| | - Matko Gulin
- Department of Radiology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Mladen Grgurević
- Department of Diabetes, Endocrinology and Metabolic Diseases Vuk Vrhovac, Merkur University Hospital, 10000 Zagreb, Croatia;
| | | |
Collapse
|
4
|
Bagnato CB, Bianco A, Bonfiglio C, Franco I, Verrelli N, Carella N, Shahini E, Zappimbulso M, Giannuzzi V, Pesole PL, Ancona A, Giannelli G. Healthy Lifestyle Changes Improve Cortisol Levels and Liver Steatosis in MASLD Patients: Results from a Randomized Clinical Trial. Nutrients 2024; 16:4225. [PMID: 39683618 PMCID: PMC11644361 DOI: 10.3390/nu16234225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Steatotic liver disease associated with metabolic dysfunction (MASLD) affects up to about 30% of the general adult population and is closely related to obesity and the metabolic syndrome. Cortisol, a stress-related hormone contributing to hepatic fat accumulation and insulin resistance, also promotes progression of the disease. The study aims to investigate the impact of lifestyle modifications on cortisol levels and hepatic steatosis in patients with MASLD. Methods: In a 16-week three-arm randomized trial, 42 patients were randomly assigned to three groups who received dietary advice (CG), dietary advice combined with aerobic exercise (AE + DA), or dietary advice with high-intensity interval training (HIIT + DA). Before the start, after 2 months of intervention, and at the end of the project, medical evaluations, routine biochemical assessments, and psychological questionnaires were analyzed. At baseline and at the end of 4 months, hepatic steatosis was evaluated by Fibroscan®. Results: In the study population, severe hepatic steatosis (74%) and obesity (98%) were prevalent at the beginning of the study. A statistically significant (p-value = 0.001) reduction in circulating cortisol levels was observed over time in the two groups doing exercise, especially in HIIT + DA (p-value = 0.006). Hepatic steatosis, assessed by Fibroscan®, disappeared in 10 participants (CAP value < 248, p-value = 0.003). CAP values and waist circumference decreased in all groups, statistically significantly in the AE + DA group (p-value = 0.005; p-value = 0.04, respectively). Conclusions: The study emphasizes the benefits of combining diet and exercise in managing MASLD. HIIT + DA significantly decreased cortisol levels, while AE + DA was the most potent intervention for reducing hepatic steatosis.
Collapse
Affiliation(s)
- Claudia Beatrice Bagnato
- Laboratory of Movement and Wellness, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (C.B.B.); (I.F.); (N.V.)
| | - Antonella Bianco
- Laboratory of Movement and Wellness, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (C.B.B.); (I.F.); (N.V.)
| | - Caterina Bonfiglio
- Data Science, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy;
| | - Isabella Franco
- Laboratory of Movement and Wellness, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (C.B.B.); (I.F.); (N.V.)
| | - Nicola Verrelli
- Laboratory of Movement and Wellness, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (C.B.B.); (I.F.); (N.V.)
| | - Nicola Carella
- Clinical Research Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy;
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (E.S.); (M.Z.); (V.G.)
| | - Marianna Zappimbulso
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (E.S.); (M.Z.); (V.G.)
| | - Vito Giannuzzi
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (E.S.); (M.Z.); (V.G.)
| | - Pasqua Letizia Pesole
- Core Facility Biobank, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (P.L.P.); (A.A.)
| | - Anna Ancona
- Core Facility Biobank, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy; (P.L.P.); (A.A.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, BA, Italy;
| |
Collapse
|
5
|
Liu Y, Mao S, Xie W, Agnieszka HLK, Helena SM, Magdalena DZ, Qian G, Ossowski Z. Relationship between physical activity and abdominal obesity and metabolic markers in postmenopausal women. Sci Rep 2024; 14:26496. [PMID: 39489777 PMCID: PMC11532536 DOI: 10.1038/s41598-024-77900-x] [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: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
This study aimed to investigate the impact of physical activity indicators monitored by the POLAR accelerometer on body obesity indicators, metabolic syndrome parameters, and energy metabolism hormones in postmenopausal women. Was included 71 participants from this study program (68.8 ± 4.3 years). We divided participants into LPA and MVPA groups based on their level of moderate to vigorous physical activity per week; Physical activity levels over 7 days were assessed using a POLAR accelerometer, with daily step counts and sedentary time recorded. Measurements included waist circumference, visceral fat volume, body fat percentage, blood pressure, fasting blood glucose, triglyceride levels, HDL cholesterol, and energy metabolism hormone levels (leptin, resistin, adiponectin). The MVPA group displayed lower waist circumference, body fat percentage, abdominal fat, and BMI compared to the LPA group (p < 0.05). A significant negative correlation was observed between daily step count and obesity indicators, including waist circumference (r = -0.301), body fat percentage (r = -0.295), abdominal fat (r = -0.318), and BMI (r = -0.238). Conversely, sedentary time showed a positive correlation with obesity indicators such as waist circumference (r = 0.258), body fat percentage (r = 0.239), and abdominal fat (r = 0.244). Moreover, daily step count exhibited a significant negative correlation with leptin levels (r = -0.245), while sedentary time was positively correlated with the energy metabolic factor leptin (r = 0.279). Waist circumference demonstrated significant positive correlations with triglycerides, blood glucose, adiponectin, resistin, and leptin levels. Postmenopausal women who engage in at least 150 min of MVPA weekly show lower obesity indices. There is a significant correlation between physical activity levels and obesity indicators, which relate to metabolic syndrome and energy metabolism factors. Thus, increased physical activity may help prevent metabolic syndrome and cardiovascular diseases in this population.
Collapse
Affiliation(s)
- Yangjun Liu
- School of Physical Education and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, People's Republic of China
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Kazimierza Górskiego 1, 80-336, Gdańsk, Poland
| | - Sujie Mao
- Discipline Construction Office of Nanjing Sport University, Nanjing, 210014, Jiangsu, People's Republic of China
| | - Wei Xie
- School of Physical Education and Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, People's Republic of China
| | | | - Sawczyn Monika Helena
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Kazimierza Górskiego 1, 80-336, Gdańsk, Poland
| | - Dzitkowska-Zabielska Magdalena
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Kazimierza Górskiego 1, 80-336, Gdańsk, Poland
| | - Guoping Qian
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Kazimierza Górskiego 1, 80-336, Gdańsk, Poland
| | - Zbigniew Ossowski
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, Kazimierza Górskiego 1, 80-336, Gdańsk, Poland.
- Gdansk University of Physical Education and Sport, Poland, 1 Kazimierza Górskiego Street, 80-336, Gdańsk, Poland.
| |
Collapse
|
6
|
Jiao W, Jiao Y, Sang Y, Wang X, Wang S. 6-Shogaol alleviates high-fat diet induced hepatic steatosis through miR-3066-5p/Grem2 pathway. Food Chem 2024; 457:140197. [PMID: 38941907 DOI: 10.1016/j.foodchem.2024.140197] [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: 04/04/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
The purpose of this study is to investigate the mechanism by which 6-shogaol ameliorates hepatic steatosis via miRNA-mRNA interaction analysis. C57BL/6 J mice were fed a high-fat diet (HFD) for 12 weeks, during which 6-shogaol was administered orally. The liver lipid level, liver function and oxidative damage in mice were evaluated. mRNA sequencing, miRNA sequencing, and RT-qPCR were employed to compare the expression profiles between the HFD group and the 6-shogaol-treated group. High-throughput sequencing was used to construct the mRNA and miRNA libraries. Target prediction and integration analysis identified eight potential miRNA-mRNA pairs involved in hepatic steatosis, which were subsequently validated in liver tissues and AML12 cells. The findings revealed that 6-shogaol modulates the miR-3066-5p/Grem2 pathway, thereby improving hepatic steatosis. This study provides new insights into the mechanisms through which 6-shogaol alleviates hepatic steatosis, establishing a foundation for future research on natural active compounds for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Wenya Jiao
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Yingshuai Jiao
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Yaxin Sang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071000, China
| | - Xianghong Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071000, China.
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
7
|
Kugler BA, Maurer A, Fu X, Franczak E, Ernst N, Schwartze K, Allen J, Li T, Crawford PA, Koch LG, Britton SL, Burgess SC, Thyfault JP. Aerobic capacity and exercise mediate protection against hepatic steatosis via enhanced bile acid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619494. [PMID: 39484384 PMCID: PMC11526936 DOI: 10.1101/2024.10.21.619494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High cardiorespiratory fitness and exercise show evidence of altering bile acid (BA) metabolism and are known to protect or treat diet-induced hepatic steatosis, respectively. Here, we tested the hypothesis that high intrinsic aerobic capacity and exercise both increase hepatic BA synthesis measured by the incorporation of 2H2O. We also leveraged mice with inducible liver-specific deletion of Cyp7a1 (LCyp7a1KO), which encodes the rate-limiting enzyme for BA synthesis, to test if exercise-induced BA synthesis is critical for exercise to reduce hepatic steatosis. The synthesis of hepatic BA, cholesterol, and de novo lipogenesis was measured in rats bred for either high (HCR) vs. low (LCR) aerobic capacity consuming acute and chronic high-fat diets. HCR rats had increased synthesis of cholesterol and certain BA species in the liver compared to LCR rats. We also found that chronic exercise with voluntary wheel running (VWR) (4 weeks) increased newly synthesized BAs of specific species in male C57BL/6J mice compared to sedentary mice. Loss of Cyp7a1 resulted in fewer new BAs and increased liver triglycerides compared to controls after a 10-week high-fat diet. Additionally, exercise via VWR for 4 weeks effectively reduced hepatic triglycerides in the high-fat diet-fed control male and female mice as expected; however, exercise in LCyp7a1KO mice did not lower liver triglycerides in either sex. These results show that aerobic capacity and exercise increase hepatic BA metabolism, which may be critical for combatting hepatic steatosis.
Collapse
Affiliation(s)
- Benjamin A. Kugler
- Departments of Cell Biology and Physiology
- Internal Medicine, Division of Endocrinology and Clinical Pharmacology and KU Diabetes Institute
| | | | - Xiaorong Fu
- Center for Human Nutrition and Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edziu Franczak
- Departments of Cell Biology and Physiology
- Internal Medicine, Division of Endocrinology and Clinical Pharmacology and KU Diabetes Institute
| | - Nick Ernst
- Departments of Cell Biology and Physiology
| | | | | | - Tiangang Li
- Department of Biochemistry and Physiology, and Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, and Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| | - Lauren G. Koch
- Dept of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | | | - Shawn C. Burgess
- Center for Human Nutrition and Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John P. Thyfault
- Departments of Cell Biology and Physiology
- Internal Medicine, Division of Endocrinology and Clinical Pharmacology and KU Diabetes Institute
- Kansas Center for Metabolism and Obesity Research, Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
8
|
Huang M, Yang J, Wang Y, Wu J. Comparative efficacy of different exercise modalities on metabolic profiles and liver functions in non-alcoholic fatty liver disease: a network meta-analysis. Front Physiol 2024; 15:1428723. [PMID: 39376897 PMCID: PMC11457013 DOI: 10.3389/fphys.2024.1428723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/28/2024] [Indexed: 10/09/2024] Open
Abstract
Objective Research evidence suggests that exercise is a potent therapeutic strategy for non-alcoholic fatty liver disease (NAFLD). Many investigations have delved into the curative potential of diverse exercise regimens on NAFLD. This investigation synthesizes findings from randomized controlled trials via a network meta-analysis to evaluate the efficacy of exercise-based interventions on NAFLD. Methods We conducted a search across five electronic databases (Web of Science, EMBASE, PubMed, SCOPUS, and CNKI)to identify randomized controlled trials (RCTs) comparing the effects of different exercise modalities on metabolic profiles and liver functions in patients with NAFLD. The literature search was comprehensive up to 15, December 2023. The selected studies were subjected to a rigorous quality appraisal and risk of bias analysis in accordance with the Cochrane Handbook's guidelines, version 5.1.0. We employed Stata/MP 17 for the network meta-analysis, presenting effect sizes as standardized mean differences (SMD). Results This study aggregated results from 28 studies, involving a total of 1,606 participants. The network meta-analysis revealed that aerobic exercise was the most effective intervention for improving BMI in patients with NAFLD, demonstrating a significant decrease in BMI (-0.72, 95%CI: -0.98 to -0.46; p < 0.05; Surface Under the Cumulative Ranking (SUCRA) = 79.8%). HIIT was the top intervention for enhancing HDL-C (0.12, 95% CI: 0.04 to 0.20; p < 0.05; SUCRA = 76.1%). Resistance exercise was the most effective for reducing LDL-C (-0.20, 95% CI: -0.33 to -0.06; p < 0.05; SUCRA = 69.7%). Mind-body exercise showed superior effectiveness in improving TC (-0.67, 95% CI: -1.10 to -0.24; p < 0.05; SUCRA = 89.7%), TG = -0.67, 95% CI: -1.10 to -0.24; p < 0.05; SUCRA = 99.6%), AST (-8.07, 95% CI: -12.88 to -3.25; p < 0.05; SUCRA = 76.1%), ALT (-12.56, 95% CI: -17.54 to -7.58; p < 0.05; SUCRA = 99.5%), and GGT (-13.77, 95% CI: -22.00 to -5.54; p < 0.05; SUCRA = 81.8%). Conclusion This network meta-analysis demonstrates that exercise interventions positively affect various metabolic profiles and liver functions in NAFLD patients. Mind-body exercises are particularly effective, surpassing other exercise forms in improving metabolic profiles and liver functions. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier registration number CRD42024526332.
Collapse
Affiliation(s)
- Mingming Huang
- School of Exercise Science and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Jiafa Yang
- School of Arts and Sports, Dong-A University, Busan, Republic of Korea
| | - Yihao Wang
- School of Exercise Science and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Jian Wu
- School of Exercise Science and Health, Capital University of Physical Education and Sports, Beijing, China
| |
Collapse
|
9
|
Chartrand DJ, Murphy-Després A, Lemieux I, Larose E, Poirier P, Després JP, Alméras N. Effects of 1,144 km of road cycling performed in 7 days: a cardiometabolic imaging study. Am J Physiol Endocrinol Metab 2024; 327:E344-E356. [PMID: 39046280 DOI: 10.1152/ajpendo.00098.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
This cardiometabolic imaging study was designed to document the adaptation of middle-aged recreational cyclists to a large exercise prescription not aiming at weight loss. Eleven middle-aged recreational male cyclists traveled 1,144 km over seven consecutive days. A comprehensive cardiometabolic profile including visceral and ectopic adiposity assessed by magnetic resonance imaging was obtained at baseline and following the exercise week. Cardiorespiratory fitness (CRF) was measured using maximal cardiopulmonary exercise testing. During the week, heart rate was monitored to calculate individual energy expenditure. Baseline characteristics of cyclists were compared with 86 healthy males in the same age range. Cyclists presented higher baseline CRF (+9.2 mL/kg/min, P < 0.0001) and lower subcutaneous (-56.2 mL, P < 0.05) and liver (-3.3%, P < 0.05) fat compared with the reference group. Despite the large energy expenditure during the cycling week, the increase in energy intake limited decreases in body weight (-0.8 ± 0.9 kg, P < 0.05) and body mass index (-0.3 ± 0.3 kg/m2, P < 0.05). Loss of fat mass (-1.5 ± 1.0 kg, P < 0.001) and a trend toward an increased lean mass (+0.8 ± 1.2 kg, P < 0.07) were observed. Visceral adiposity (-14.1 ± 14.2 mL, P < 0.01) and waist circumference (-3.2 ± 1.7 cm, P < 0.0001) decreased, whereas subcutaneous (-2.7 ± 5.1 mL, NS), liver (-0.5 ± 0.9%, NS), and cardiac (-0.3 ± 2.3 mL, NS) fat remained unchanged. This cardiometabolic imaging study documents middle-aged recreational cyclists' subcutaneous and visceral adiposity as well as cardiac and liver fat responses to a large volume of endurance exercise despite an increase in energy intake aimed at limiting weight loss.NEW & NOTEWORTHY Even when being accompanied by a substantial increase in energy intake to compensate energy expenditure and limit weight loss, a large volume of endurance exercise performed within a short period of time is associated with a significant reduction in visceral adiposity. High cardiorespiratory fitness is associated with low levels of liver fat in middle-aged males.
Collapse
Affiliation(s)
- Dominic J Chartrand
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Adrien Murphy-Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Isabelle Lemieux
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
| | - Eric Larose
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| | - Paul Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
- Faculty of Pharmacy, Université Laval, Québec, Québec, Canada
| | - Jean-Pierre Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
- VITAM-Centre de recherche en santé durable, CIUSSS de la Capitale-Nationale, Québec, Québec, Canada
| | - Natalie Alméras
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Québec, Québec, Canada
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Québec, Canada
| |
Collapse
|
10
|
Esteves JV, Stanford KI. Exercise as a tool to mitigate metabolic disease. Am J Physiol Cell Physiol 2024; 327:C587-C598. [PMID: 38981607 PMCID: PMC11427015 DOI: 10.1152/ajpcell.00144.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Metabolic diseases, notably obesity and type 2 diabetes (T2D), have reached alarming proportions and constitute a significant global health challenge, emphasizing the urgent need for effective preventive and therapeutic strategies. In contrast, exercise training emerges as a potent intervention, exerting numerous positive effects on metabolic health through adaptations to the metabolic tissues. Here, we reviewed the major features of our current understanding with respect to the intricate interplay between metabolic diseases and key metabolic tissues, including adipose tissue, skeletal muscle, and liver, describing some of the main underlying mechanisms driving pathogenesis, as well as the role of exercise to combat and treat obesity and metabolic disease.
Collapse
Affiliation(s)
- Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
11
|
Li DD, Yang X, Yang Y, Zhao LH, Zhang TT, Wang YN, Su JB, Wang LH, Shi HY. Association of Questionnaire-Based Physical Activity Analysis and Body Composition Dynamics in Type 2 Diabetes: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2024; 17:2955-2966. [PMID: 39135891 PMCID: PMC11318602 DOI: 10.2147/dmso.s459356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
Background Physical activity (PA) exerts an important influence on glycemic control in type 2 diabetes (T2D) patients. Alterations in body composition in patients with T2D may be involved in the overall pathophysiologic process, but PAs and alterations in body composition have been poorly studied. Methods A total of 615 patients with T2D were selected by convenient sampling. The patients were investigated with the International Physical Activity Questionnaire (IPAQ-S). Moreover, biochemical indices were collected, and the progression of the body composition of the subjects was determined via dual-energy X-ray absorptiometry (DXA). The variables included lumbar bone mineral density (LSBMD), femoral neck bone mineral density (FNBMD), hip bone mineral density (HBMD), whole-body bone mineral density (TBMD), limb skeletal muscle mass index (ASMI), whole-body fat percentage (B-FAT) and trunk fat percentage (T-FAT). Moreover, the levels of physical activity (high level of physical activity [H-PA], medium level of physical activity [M-PA] and low level of physical activity [L-PA]) were divided into three groups to analyze the changes in patient body composition with changes in physical activity level. Results One-way analysis of variance showed that β-CTX, TP1NP, HbA1c, B-FAT and T-FAT increased significantly (p<0.05), while 25(OH)D, LSBMD, FNBMD, HBMD, TBMD and ASMI decreased significantly (p<0.001) with the decrease of physical activity. However, there was no significant difference in serum lipids between lnHOMA-ir and lnHOMA-β (p>0.05). Multiple linear regression model was established to gradually adjust for clinical confounding factors. It was found that physical activity level was independently positively correlated with LSBMD, FNBMD, HBMD, TBMD, and ASMI, and was independently negatively correlated with B-FAT and T-FAT in patients with type 2 diabetes. Conclusion A lack of physical activity is an independent risk factor for decreased bone mineral density, decreased skeletal muscle content and increased fat content in patients with T2D.
Collapse
Affiliation(s)
- Dan-Dan Li
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Xue Yang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Yang Yang
- Department of General Practice, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Li-Hua Zhao
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Tian-Tian Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Yi-Nan Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Jian-Bin Su
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Li-Hua Wang
- Department of Nursing, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| | - Hai-Yan Shi
- Department of Hepatobiliary Surgery, Affiliated Hospital 2 of Nantong University, and First People’s Hospital of Nantong City, Nantong, 226006, People’s Republic of China
| |
Collapse
|
12
|
Amar D, Gay NR, Jean-Beltran PM, Bae D, Dasari S, Dennis C, Evans CR, Gaul DA, Ilkayeva O, Ivanova AA, Kachman MT, Keshishian H, Lanza IR, Lira AC, Muehlbauer MJ, Nair VD, Piehowski PD, Rooney JL, Smith KS, Stowe CL, Zhao B, Clark NM, Jimenez-Morales D, Lindholm ME, Many GM, Sanford JA, Smith GR, Vetr NG, Zhang T, Almagro Armenteros JJ, Avila-Pacheco J, Bararpour N, Ge Y, Hou Z, Marwaha S, Presby DM, Natarajan Raja A, Savage EM, Steep A, Sun Y, Wu S, Zhen J, Bodine SC, Esser KA, Goodyear LJ, Schenk S, Montgomery SB, Fernández FM, Sealfon SC, Snyder MP, Adkins JN, Ashley E, Burant CF, Carr SA, Clish CB, Cutter G, Gerszten RE, Kraus WE, Li JZ, Miller ME, Nair KS, Newgard C, Ortlund EA, Qian WJ, Tracy R, Walsh MJ, Wheeler MT, Dalton KP, Hastie T, Hershman SG, Samdarshi M, Teng C, Tibshirani R, Cornell E, Gagne N, May S, Bouverat B, Leeuwenburgh C, Lu CJ, Pahor M, Hsu FC, Rushing S, Walkup MP, Nicklas B, Rejeski WJ, Williams JP, Xia A, Albertson BG, Barton ER, Booth FW, Caputo T, Cicha M, De Sousa LGO, Farrar R, Hevener AL, Hirshman MF, Jackson BE, Ke BG, Kramer KS, Lessard SJ, Makarewicz NS, Marshall AG, Nigro P, Powers S, Ramachandran K, Rector RS, Richards CZT, Thyfault J, Yan Z, Zang C, Amper MAS, Balci AT, Chavez C, Chikina M, Chiu R, Gritsenko MA, Guevara K, Hansen JR, Hennig KM, Hung CJ, Hutchinson-Bunch C, Jin CA, Liu X, Maner-Smith KM, Mani DR, Marjanovic N, Monroe ME, Moore RJ, Moore SG, Mundorff CC, Nachun D, Nestor MD, Nudelman G, Pearce C, Petyuk VA, Pincas H, Ramos I, Raskind A, Rirak S, Robbins JM, Rubenstein AB, Ruf-Zamojski F, Sagendorf TJ, Seenarine N, Soni T, Uppal K, Vangeti S, Vasoya M, Vornholt A, Yu X, Zaslavsky E, Zebarjadi N, Bamman M, Bergman BC, Bessesen DH, Buford TW, Chambers TL, Coen PM, Cooper D, Haddad F, Gadde K, Goodpaster BH, Harris M, Huffman KM, Jankowski CM, Johannsen NM, Kohrt WM, Lester B, Melanson EL, Moreau KL, Musi N, Newton RL, Radom-Aizik S, Ramaker ME, Rankinen T, Rasmussen BB, Ravussin E, Schauer IE, Schwartz RS, Sparks LM, Thalacker-Mercer A, Trappe S, Trappe TA, Volpi E. Temporal dynamics of the multi-omic response to endurance exercise training. Nature 2024; 629:174-183. [PMID: 38693412 PMCID: PMC11062907 DOI: 10.1038/s41586-023-06877-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/16/2023] [Indexed: 05/03/2024]
Abstract
Regular exercise promotes whole-body health and prevents disease, but the underlying molecular mechanisms are incompletely understood1-3. Here, the Molecular Transducers of Physical Activity Consortium4 profiled the temporal transcriptome, proteome, metabolome, lipidome, phosphoproteome, acetylproteome, ubiquitylproteome, epigenome and immunome in whole blood, plasma and 18 solid tissues in male and female Rattus norvegicus over eight weeks of endurance exercise training. The resulting data compendium encompasses 9,466 assays across 19 tissues, 25 molecular platforms and 4 training time points. Thousands of shared and tissue-specific molecular alterations were identified, with sex differences found in multiple tissues. Temporal multi-omic and multi-tissue analyses revealed expansive biological insights into the adaptive responses to endurance training, including widespread regulation of immune, metabolic, stress response and mitochondrial pathways. Many changes were relevant to human health, including non-alcoholic fatty liver disease, inflammatory bowel disease, cardiovascular health and tissue injury and recovery. The data and analyses presented in this study will serve as valuable resources for understanding and exploring the multi-tissue molecular effects of endurance training and are provided in a public repository ( https://motrpac-data.org/ ).
Collapse
|
13
|
Chen J, Luo Q, Su Y, Wang J, Fang Z, Luo F. Effects of physical activity on the levels of remnant cholesterol: A population-based study. J Cell Mol Med 2024; 28:e18062. [PMID: 38018906 PMCID: PMC10844695 DOI: 10.1111/jcmm.18062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/05/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023] Open
Abstract
Physical activity (PA) has the potential to bring about favourable changes in plasma lipid profile. However, the relationship between PA and remnant cholesterol (RC) remains unclear. We aimed to study the link between PA and RC using the database of the 2007-2020 National Health and Nutrition Examination Survey (NHANES). PA was categorized based on Physical Activity Guidelines for Americans. A multivariate linear regression model was used to determine the correlations between PA and RC. The study involved a total of 18,396 participants and revealed that individuals whose PA met the guidelines by engaging in moderate-intensity PA at least 150 min per week had lower body mass index and showed decreased levels of triglyceride, TC, and haemoglobin A1c compared to those who were physically inactive, exercising <150 min per week. Participants whose intensity of PA meets PA guidelines had a lower level of RC than those who did not met PA guidelines (β = -1.3, 95% confidence interval [CI]: -1.9 to -0.7, p < 0.001), even after adjusting for confounders. During subgroup analysis, we observed that race (pinteraction = 0.0089) emerged as a significant factor of interaction.
Collapse
Affiliation(s)
- Jingfei Chen
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Qin Luo
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of Cardiovascular Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yingjie Su
- Department of Emergency Medicine, The Affiliated Changsha Central Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunanChina
| | - Jiangang Wang
- Department of Health Management, The Third Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zhenfei Fang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of Cardiovascular Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Fei Luo
- Research Institute of Blood Lipid and Atherosclerosis, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Department of Cardiovascular Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
14
|
Hughey CC, Bracy DP, Rome FI, Goelzer M, Donahue EP, Viollet B, Foretz M, Wasserman DH. Exercise training adaptations in liver glycogen and glycerolipids require hepatic AMP-activated protein kinase in mice. Am J Physiol Endocrinol Metab 2024; 326:E14-E28. [PMID: 37938177 PMCID: PMC11193517 DOI: 10.1152/ajpendo.00289.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/09/2023]
Abstract
Regular exercise elicits adaptations in glucose and lipid metabolism that allow the body to meet energy demands of subsequent exercise bouts more effectively and mitigate metabolic diseases including fatty liver. Energy discharged during the acute exercise bouts that comprise exercise training may be a catalyst for liver adaptations. During acute exercise, liver glycogenolysis and gluconeogenesis are accelerated to supply glucose to working muscle. Lower liver energy state imposed by gluconeogenesis and related pathways activates AMP-activated protein kinase (AMPK), which conserves ATP partly by promoting lipid oxidation. This study tested the hypothesis that AMPK is necessary for liver glucose and lipid adaptations to training. Liver-specific AMPKα1α2 knockout (AMPKα1α2fl/fl+AlbCre) mice and littermate controls (AMPKα1α2fl/fl) completed sedentary and exercise training protocols. Liver nutrient fluxes were quantified at rest or during acute exercise following training. Liver metabolites and molecular regulators of metabolism were assessed. Training increased liver glycogen in AMPKα1α2fl/fl mice, but not in AMPKα1α2fl/fl+AlbCre mice. The inability to increase glycogen led to lower glycogenolysis, glucose production, and circulating glucose during acute exercise in trained AMPKα1α2fl/fl+AlbCre mice. Deletion of AMPKα1α2 attenuated training-induced declines in liver diacylglycerides. In particular, training lowered the concentration of unsaturated and elongated fatty acids comprising diacylglycerides in AMPKα1α2fl/fl mice, but not in AMPKα1α2fl/fl+AlbCre mice. Training increased liver triacylglycerides and the desaturation and elongation of fatty acids in triacylglycerides of AMPKα1α2fl/fl+AlbCre mice. These lipid responses were independent of differences in tricarboxylic acid cycle fluxes. In conclusion, AMPK is required for liver training adaptations that are critical to glucose and lipid metabolism.NEW & NOTEWORTHY This study shows that the energy sensor and transducer, AMP-activated protein kinase (AMPK), is necessary for an exercise training-induced: 1) increase in liver glycogen that is necessary for accelerated glycogenolysis during exercise, 2) decrease in liver glycerolipids independent of tricarboxylic acid (TCA) cycle flux, and 3) decline in the desaturation and elongation of fatty acids comprising liver diacylglycerides. The mechanisms defined in these studies have implications for use of regular exercise or AMPK-activators in patients with fatty liver.
Collapse
Affiliation(s)
- Curtis C Hughey
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Deanna P Bracy
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, United States
| | - Ferrol I Rome
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Mickael Goelzer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - E Patrick Donahue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benoit Viollet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Marc Foretz
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
15
|
Zhang M, Xiao B, Chen X, Ou B, Wang S. Physical exercise plays a role in rebalancing the bile acids of enterohepatic axis in non-alcoholic fatty liver disease. Acta Physiol (Oxf) 2024; 240:e14065. [PMID: 38037846 DOI: 10.1111/apha.14065] [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: 05/26/2023] [Revised: 10/09/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered as one of the most common diseases of lipid metabolism disorders, which is closely related to bile acids disorders and gut microbiota disorders. Bile acids are synthesized from cholesterol in the liver, and processed by gut microbiota in intestinal tract, and participate in metabolic regulation through the enterohepatic circulation. Bile acids not only promote the consumption and absorption of intestinal fat but also play an important role in biological metabolic signaling network, affecting fat metabolism and glucose metabolism. Studies have demonstrated that exercise plays an important role in regulating the composition and function of bile acid pool in enterohepatic axis, which maintains the homeostasis of the enterohepatic circulation and the health of the host gut microbiota. Exercise has been recommended by several health guidelines as the first-line intervention for patients with NAFLD. Can exercise alter bile acids through the microbiota in the enterohepatic axis? If so, regulating bile acids through exercise may be a promising treatment strategy for NAFLD. However, the specific mechanisms underlying this potential connection are largely unknown. Therefore, in this review, we tried to review the relationship among NAFLD, physical exercise, bile acids, and gut microbiota through the existing data and literature, highlighting the role of physical exercise in rebalancing bile acid and microbial dysbiosis.
Collapse
Affiliation(s)
- Minyu Zhang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Biyang Xiao
- College of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Xiaoqi Chen
- College of Life Sciences, Zhaoqing University, Zhaoqing, China
| | - Bingming Ou
- College of Life Sciences, Zhaoqing University, Zhaoqing, China
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Songtao Wang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| |
Collapse
|
16
|
Damasceno de Lima R, Fudoli Lins Vieira R, Rosetto Muñoz V, Chaix A, Azevedo Macedo AP, Calheiros Antunes G, Felonato M, Rosseto Braga R, Castelo Branco Ramos Nakandakari S, Calais Gaspar R, Ramos da Silva AS, Esper Cintra D, Pereira de Moura L, Mekary RA, Rochete Ropelle E, Pauli JR. Time-restricted feeding combined with resistance exercise prevents obesity and improves lipid metabolism in the liver of mice fed a high-fat diet. Am J Physiol Endocrinol Metab 2023; 325:E513-E528. [PMID: 37755454 PMCID: PMC10864020 DOI: 10.1152/ajpendo.00129.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a condition characterized by the accumulation of fat in the liver, is estimated to be the most common liver disease worldwide. Obesity is a major risk factor and contributor, and, accordingly, weight loss can improve NAFLD. Previous studies in preclinical models of diet-induced obesity and fatty liver disease have shown the independent benefits of resistance exercise training (RT) and time-restricted feeding (TRF) in preventing weight gain and hepatic build-up of fat. Here, we tested the combined effect of TRF and RT on obesity and NAFLD in mice fed a high-fat diet. Our results showed that both TRF-8-h food access in the active phase-and RT-consisting of three weekly sessions of ladder climbing-attenuated body weight gain, improved glycemic homeostasis, and decreased the accumulation of lipids in the liver. TRF combined with RT improved the respiratory exchange rate, energy expenditure, and mitochondrial respiration in the liver. Furthermore, gene expression analysis in the liver revealed lower mRNA expression of lipogenesis and inflammation genes along with increased mRNA of fatty acid oxidation genes in the TRF + RT group. Importantly, combined TRF + RT was shown to be more efficient in preventing obesity and metabolic disorders. In conclusion, TRF and RT exert complementary actions compared with isolated interventions, with significant effects on metabolic disorders and NAFLD in mice.NEW & NOTEWORTHY Whether time-restricted feeding (TRF) combined with resistance exercise training (RT) may be more efficient compared with these interventions alone is still unclear. We show that when combined with RT, TRF provided additional benefits, being more effective in increasing energy expenditure, preventing weight gain, and regulating glycemic homeostasis than each intervention alone. Thus, our results demonstrate that TRF and RT have complementary actions on some synergistic pathways that prevented obesity and hepatic liver accumulation.
Collapse
Affiliation(s)
- Robson Damasceno de Lima
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, United States
| | - Ana Paula Azevedo Macedo
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Gabriel Calheiros Antunes
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Maíra Felonato
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Renata Rosseto Braga
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | | | - Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
| | - Adelino Sanchez Ramos da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, and Postgraduate Program in Physical Education and Sport, School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics (LabGeN), University of Campinas (UNICAMP), Limeira, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| | - Rania A Mekary
- Massachusetts College of Pharmacy and Health Sciences (MCPHS) University, Boston, Massachusetts, United States
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| |
Collapse
|
17
|
Tan L, Yan W, Yang W, Kamionka A, Lipowski M, Zhao Z, Zhao G. Effect of exercise on inflammatory markers in postmenopausal women with overweight and obesity: A systematic review and meta-analysis. Exp Gerontol 2023; 183:112310. [PMID: 37844768 DOI: 10.1016/j.exger.2023.112310] [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: 07/10/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
IMPORTANCE Postmenopausal women affected by overweight and obesity are susceptible to a variety of diseases due to inflammation. Exercise may reduce the risk of disease by attenuating low-grade chronic inflammation. OBJECTIVE We conducted a systematic review and meta-analysis to investigate the effects of exercise on inflammatory markers in postmenopausal women struggling with overweight and obesity. METHOD Literature as of May 2023 was searched from databases such as Cochrane, Embase, Pubmed, Web of Science, and EBSCO and English-language randomized controlled trials (RCTs) that meet the inclusion criteria were selected. Studies were included based on the following criteria: (A) Written in English; (B) RCTs; (C) Postmenopausal women impacted by overweight and obesity as research objects; (D) Outcome measurements include CRP, TNF-α, IL-6, and adiponectin; (E) Duration of the exercise intervention is eight weeks. RESULTS A total of 34 articles and 2229 participants were included. Exercise can significantly reduce the level of C-reactive protein (CRP) (MD: -0.59, 95 % CI: -0.87 to -0.31, p < 0.00001), tumor necrosis factor-α (TNF-α) (MD: -0.65, 95 % CI: -0.94 to -0.35, p < 0.00001), interleukin-6 (IL-6) (MD: -0.48, 95 % CI: -0.75 to -0.21, p < 0.00001), and exercise can significantly increase the level of adiponectin (MD: 0.33, 95 % CI: 0.02 to 0.65, p = 0.04) in women impacted by overweight and obesity. CONCLUSION These results suggest that exercise may be an effective intervention for reducing pro-inflammatory markers and increasing adiponectin in postmenopausal women impacted by overweight and obesity. The findings may provide clinicians and healthcare professionals with insights into the implementation of exercise programs for postmenopausal women living with overweight and obesity.
Collapse
Affiliation(s)
- Liang Tan
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; Changsha Commerce & Tourism College, 410116, Changsha, China.
| | - Weihua Yan
- School of Management, Beijing Sport University, 100084, Beijing, China.
| | - Weilin Yang
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland.
| | - Agata Kamionka
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland.
| | - Mariusz Lipowski
- Faculty of Social and Humanities, University WSB Merito, 80-266 Gdansk, Poland.
| | - Zijian Zhao
- Physical Education Institute (Main Campus), Zhengzhou University, 450001, Zhengzhou, China.
| | - Gang Zhao
- Changsha Commerce & Tourism College, 410116, Changsha, China.
| |
Collapse
|
18
|
Igudesman D, Mucinski J, Harrison S, Cawthon PM, Linge J, Goodpaster BH, Cummings SR, Hepple RT, Jurczak MJ, Kritchevsky SB, Marcinek D, Coen PM, Corbin KD. Associations of Skeletal Muscle Mass, Muscle Fat Infiltration, Mitochondrial Energetics, and Cardiorespiratory Fitness with Liver Fat Among Older Adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.24.23297480. [PMID: 37961367 PMCID: PMC10635187 DOI: 10.1101/2023.10.24.23297480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Muscle mass loss may be associated with liver fat accumulation, yet scientific consensus is lacking and evidence in older adults is scant. It is unclear which muscle characteristics might contribute to this association in older adults. Methods We associated comprehensive muscle-related phenotypes including muscle mass normalized to body weight (D 3 -creatine dilution), muscle fat infiltration (MRI), carbohydrate-supported muscle mitochondrial maximal oxidative phosphorylation (respirometry), and cardiorespiratory fitness (VO 2 peak) with liver fat among older adults. Linear regression models adjusted for age, gender, technician (respirometry only), daily minutes of moderate to vigorous physical activity, and prediabetes/diabetes status tested main effects and interactions of each independent variable with waist circumference (high: women-≥88 cm, men-≥102 cm) and gender. Results Among older adults aged 75 (IQR 73, 79 years; 59.8% women), muscle mass and liver fat were not associated overall but were positively associated among participants with a high waist circumference (β: 25.2; 95%CI 11.7, 40.4; p =.0002; N=362). Muscle fat infiltration and liver fat were positively associated (β: 15.2; 95%CI 6.8, 24.3; p =.0003; N=378). Carbohydrate-supported maximum oxidative phosphorylation and VO 2 peak (adjusted β: -12.9; 95%CI -20.3, -4.8; p =0.003; N=361) were inversely associated with liver fat; adjustment attenuated the estimate for maximum oxidative phosphorylation although the point estimate remained negative (β: -4.0; 95%CI -11.6, 4.2; p =0.32; N=321). Conclusions Skeletal muscle-related characteristics are metabolically relevant factors linked to liver fat in older adults. Future research should confirm our results to determine whether trials targeting mechanisms common to liver and muscle fat accumulation are warranted.
Collapse
|
19
|
Moore TM, Lee S, Olsen T, Morselli M, Strumwasser AR, Lin AJ, Zhou Z, Abrishami A, Garcia SM, Bribiesca J, Cory K, Whitney K, Ho T, Ho T, Lee JL, Rucker DH, Nguyen CQA, Anand ATS, Yackly A, Mendoza LQ, Leyva BK, Aliman C, Artiga DJ, Meng Y, Charugundla S, Pan C, Jedian V, Seldin MM, Ahn IS, Diamante G, Blencowe M, Yang X, Mouisel E, Pellegrini M, Turcotte LP, Birkeland KI, Norheim F, Drevon CA, Lusis AJ, Hevener AL. Conserved multi-tissue transcriptomic adaptations to exercise training in humans and mice. Cell Rep 2023; 42:112499. [PMID: 37178122 PMCID: PMC11352395 DOI: 10.1016/j.celrep.2023.112499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/04/2022] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Physical activity is associated with beneficial adaptations in human and rodent metabolism. We studied over 50 complex traits before and after exercise intervention in middle-aged men and a panel of 100 diverse strains of female mice. Candidate gene analyses in three brain regions, muscle, liver, heart, and adipose tissue of mice indicate genetic drivers of clinically relevant traits, including volitional exercise volume, muscle metabolism, adiposity, and hepatic lipids. Although ∼33% of genes differentially expressed in skeletal muscle following the exercise intervention are similar in mice and humans independent of BMI, responsiveness of adipose tissue to exercise-stimulated weight loss appears controlled by species and underlying genotype. We leveraged genetic diversity to generate prediction models of metabolic trait responsiveness to volitional activity offering a framework for advancing personalized exercise prescription. The human and mouse data are publicly available via a user-friendly Web-based application to enhance data mining and hypothesis development.
Collapse
Affiliation(s)
- Timothy M Moore
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA; Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Sindre Lee
- Department of Transplantation, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA; UCLA-DOE Institute for Genomics and Proteomics, University of California Los Angeles, Los Angeles, CA, USA; Institute for Quantitative and Computational Biosciences - The Collaboratory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexander R Strumwasser
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Amanda J Lin
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA; Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford, CA, USA
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Aaron Abrishami
- Department of Transplantation, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Steven M Garcia
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Jennifer Bribiesca
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Kevin Cory
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Kate Whitney
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Theodore Ho
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Timothy Ho
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Joseph L Lee
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel H Rucker
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Christina Q A Nguyen
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Akshay T S Anand
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Aidan Yackly
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Lorna Q Mendoza
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Brayden K Leyva
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Claudia Aliman
- Department of Transplantation, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Daniel J Artiga
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Yonghong Meng
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Sarada Charugundla
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Calvin Pan
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Vida Jedian
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA
| | - Marcus M Seldin
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA; Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - In Sook Ahn
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Graciel Diamante
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Montgomery Blencowe
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Etienne Mouisel
- Institute of Metabolic and Cardiovascular Diseases, UMR1297 Inserm, Paul Sabatier University, Toulouse, France
| | - Matteo Pellegrini
- UCLA-DOE Institute for Genomics and Proteomics, University of California Los Angeles, Los Angeles, CA, USA
| | - Lorraine P Turcotte
- Department of Biological Sciences, Dana & David Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Kåre I Birkeland
- Department of Transplantation, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Frode Norheim
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA; Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Aldons J Lusis
- Division of Cardiology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, CA, USA; Iris Cantor-UCLA Women's Health Research Center, Los Angeles, CA, USA; Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Los Angeles, CA, USA.
| |
Collapse
|
20
|
Muñoz VR, Gaspar RC, Mancini MCS, de Lima RD, Vieira RFL, Crisol BM, Antunes GC, Trombeta JCS, Bonfante ILP, Simabuco FM, da Silva ASR, Cavaglieri CR, Ropelle ER, Cintra DE, Pauli JR. Short-term physical exercise controls age-related hyperinsulinemia and improves hepatic metabolism in aged rodents. J Endocrinol Invest 2023; 46:815-827. [PMID: 36318449 DOI: 10.1007/s40618-022-01947-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/19/2022] [Indexed: 03/18/2023]
Abstract
PURPOSE Aging is associated with changes in glucose homeostasis related to both decreased insulin secretion and/or impaired insulin action, contributing to the high prevalence of type 2 diabetes (T2D) in the elderly population. Additionally, studies are showing that chronically high levels of circulating insulin can also lead to insulin resistance. In contrast, physical exercise has been a strategy used to improve insulin sensitivity and metabolic health. However, the molecular alterations resulting from the effects of physical exercise in the liver on age-related hyperinsulinemia conditions are not yet fully established. This study aimed to investigate the effects of 7 days of aerobic exercise on hepatic metabolism in aged hyperinsulinemic rats (i.e., Wistar and F344) and in Slc2a4+/- mice (hyperglycemic and hyperinsulinemic mice). RESULTS Both aged models showed alterations in insulin and glucose tolerance, which were associated with essential changes in hepatic fat metabolism (lipogenesis, gluconeogenesis, and inflammation). In contrast, 7 days of physical exercise was efficient in improving whole-body glucose and insulin sensitivity, and hepatic metabolism. The Slc2a4+/- mice presented significant metabolic impairments (insulin resistance and hepatic fat accumulation) that were improved by short-term exercise training. In this scenario, high circulating insulin may be an important contributor to age-related insulin resistance and hepatic disarrangements in some specific conditions. CONCLUSION In conclusion, our data demonstrated that short-term aerobic exercise was able to control mechanisms related to hepatic fat accumulation and insulin sensitivity in aged rodents. These effects could contribute to late-life metabolic health and prevent the development/progression of age-related T2D.
Collapse
Affiliation(s)
- V R Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - R C Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - M C S Mancini
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - R D de Lima
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - R F L Vieira
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - B M Crisol
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - G C Antunes
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - J C S Trombeta
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil
| | - I L P Bonfante
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil
| | - F M Simabuco
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - A S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - C R Cavaglieri
- Exercise Physiology Laboratory (FISEX), Faculty of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil
| | - E R Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- National Institute of Science and Technology of Obesity and Diabetes, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - D E Cintra
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - J R Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
- National Institute of Science and Technology of Obesity and Diabetes, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
21
|
Zhang T, Tian J, Fan J, Liu X, Wang R. Exercise training-attenuated insulin resistance and liver injury in elderly pre-diabetic patients correlates with NLRP3 inflammasome. Front Immunol 2023; 14:1082050. [PMID: 36817440 PMCID: PMC9929576 DOI: 10.3389/fimmu.2023.1082050] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Background Diabetes is one of the most common metabolic diseases and continues to be a leading cause of death worldwide. The NLRP3 inflammasome has been shown to exert detrimental effects on diabetic models. However, evidence linking NLRP3 inflammasome and pre-diabetes has been scarcely explored. Herein, we aimed to determine whether the NLRP3 inflammasome correlates with insulin resistance and liver pathology in a cohort of pre-diabetic subjects. Methods 50 pre-diabetic subjects were randomly assigned to a Pre-diabetes Control (DC, n=25) and a Pre-diabetes exercise (DEx, n=25) group. 25 Normal subjects (NC) were selected as controls. The DEx group performed a 6-month combined Yijingjing and resistance training intervention, while DC and NC group remained daily routines. Clinical metabolic parameters were determined with an automatic biochemistry analyzer; inflammatory cytokines were quantified by the ELISA assay; the protein expressions of NLRP3 inflammasome components in PBMCs were evaluated by Western Blot. Results The insulin resistance, liver injury and NLRP3 inflammasome activity were higher in pre-diabetic individuals than in normal control group. However, 6-month exercise intervention counteracted this trend, significantly improved insulin sensitivity, reduced liver injury and inhibited the overactivation of NLRP3 inflammasome in pre-diabetic subjects. Moreover, positive correlations between insulin resistance, liver pathology and NLRP3 inflammasome were also found. Conclusions Our study suggests that exercise training is an effective strategy to alleviate insulin resistance and liver injury in elderly pre-diabetic subjects which is probably associated with the inhibition of NLRP3 inflammasome activity.
Collapse
Affiliation(s)
- Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Jingjing Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Jingcheng Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Xiangyun Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
22
|
Flack KD, Vítek L, Fry CS, Stec DE, Hinds TD. Cutting edge concepts: Does bilirubin enhance exercise performance? Front Sports Act Living 2023; 4:1040687. [PMID: 36713945 PMCID: PMC9874874 DOI: 10.3389/fspor.2022.1040687] [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] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Exercise performance is dependent on many factors, such as muscular strength and endurance, cardiovascular capacity, liver health, and metabolic flexibility. Recent studies show that plasma levels of bilirubin, which has classically been viewed as a liver dysfunction biomarker, are elevated by exercise training and that elite athletes may have significantly higher levels. Other studies have shown higher plasma bilirubin levels in athletes and active individuals compared to general, sedentary populations. The reason for these adaptions is unclear, but it could be related to bilirubin's antioxidant properties in response to a large number of reactive oxygen species (ROS) that originates from mitochondria during exercise. However, the mechanisms of these are unknown. Current research has re-defined bilirubin as a metabolic hormone that interacts with nuclear receptors to drive gene transcription, which reduces body weight. Bilirubin has been shown to reduce adiposity and improve the cardiovascular system, which might be related to the adaption of bilirubin increasing during exercise. No studies have directly tested if elevating bilirubin levels can influence athletic performance. However, based on the mechanisms proposed in the present review, this seems plausible and an area to consider for future studies. Here, we discuss the importance of bilirubin and exercise and how the combination might improve metabolic health outcomes and possibly athletic performance.
Collapse
Affiliation(s)
- Kyle D. Flack
- Department of Dietetics and Human Nutrition, University of Kentucky, Lexington, KY, United States,Correspondence: Kyle D. Flack Terry D. Hinds
| | - Libor Vítek
- 4th Department of Internal Medicine and Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Christopher S. Fry
- Department of Athletic Training and Clinical Nutrition, University of Kentucky College of Medicine, Lexington, KY, United States,Center for Muscle Biology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - David E. Stec
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States,Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, KY, United States,Markey Cancer Center, University of Kentucky, Lexington, KY, United States,Correspondence: Kyle D. Flack Terry D. Hinds
| |
Collapse
|
23
|
Ezpeleta M, Gabel K, Cienfuegos S, Kalam F, Lin S, Pavlou V, Song Z, Haus JM, Koppe S, Alexandria SJ, Tussing-Humphreys L, Varady KA. Effect of alternate day fasting combined with aerobic exercise on non-alcoholic fatty liver disease: A randomized controlled trial. Cell Metab 2023; 35:56-70.e3. [PMID: 36549296 PMCID: PMC9812925 DOI: 10.1016/j.cmet.2022.12.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/20/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
Innovative non-pharmacological lifestyle strategies to treat non-alcoholic fatty liver disease (NAFLD) are critically needed. This study compared the effects of alternate day fasting (ADF) combined with exercise to fasting alone, or exercise alone, on intrahepatic triglyceride (IHTG) content. Adults with obesity and NAFLD (n = 80, 81% female, age: 23-65 years) were randomized to 1 of 4 groups for 3 months: combination of ADF (600 kcal/2,500 kJ "fast day" alternated with an ad libitum intake "feast day") and moderate-intensity aerobic exercise (5 session per week, 60 min/session); ADF alone; exercise alone; or a no-intervention control group. By month 3, IHTG content was significantly reduced in the combination group (-5.48%; 95% CI, -7.77% to -3.18%), compared with the exercise group (-1.30%; 95% CI, -3.80% to 1.20%; p = 0.02) and the control group (-0.17%; 95% CI, -2.17% to 1.83%; p < 0.01) but was not significantly different versus the ADF group (-2.25%; 95% CI, -4.46% to -0.04%; p = 0.05). Body weight, fat mass, waist circumference, and alanine transaminase (ALT) levels significantly decreased, while insulin sensitivity significantly increased in the combination group compared with the control group. Lean mass, aspartate transaminase (AST), HbA1c, blood pressure, plasma lipids, liver fibrosis score, and hepatokines (fetuin-A, FGF-21, and selenoprotein P) did not differ between groups. Combining intermittent fasting with exercise is effective for reducing hepatic steatosis in patients with NAFLD but may offer no additional benefit versus fasting alone.
Collapse
Affiliation(s)
- Mark Ezpeleta
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Kelsey Gabel
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Sofia Cienfuegos
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Faiza Kalam
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Shuhao Lin
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Vasiliki Pavlou
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Jacob M Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Sean Koppe
- Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shaina J Alexandria
- Department of Preventative Medicine (Biostatistics), Northwestern University, Chicago, IL, USA
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Krista A Varady
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
24
|
Rome FI, Shobert GL, Voigt WC, Stagg DB, Puchalska P, Burgess SC, Crawford PA, Hughey CC. Loss of hepatic phosphoenolpyruvate carboxykinase 1 dysregulates metabolic responses to acute exercise but enhances adaptations to exercise training in mice. Am J Physiol Endocrinol Metab 2023; 324:E9-E23. [PMID: 36351254 PMCID: PMC9799143 DOI: 10.1152/ajpendo.00222.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Acute exercise increases liver gluconeogenesis to supply glucose to working muscles. Concurrently, elevated liver lipid breakdown fuels the high energetic cost of gluconeogenesis. This functional coupling between liver gluconeogenesis and lipid oxidation has been proposed to underlie the ability of regular exercise to enhance liver mitochondrial oxidative metabolism and decrease liver steatosis in individuals with nonalcoholic fatty liver disease. Herein we tested whether repeated bouts of increased hepatic gluconeogenesis are necessary for exercise training to lower liver lipids. Experiments used diet-induced obese mice lacking hepatic phosphoenolpyruvate carboxykinase 1 (KO) to inhibit gluconeogenesis and wild-type (WT) littermates. 2H/13C metabolic flux analysis quantified glucose and mitochondrial oxidative fluxes in untrained mice at rest and during acute exercise. Circulating and tissue metabolite levels were determined during sedentary conditions, acute exercise, and refeeding postexercise. Mice also underwent 6 wk of treadmill running protocols to define hepatic and extrahepatic adaptations to exercise training. Untrained KO mice were unable to maintain euglycemia during acute exercise resulting from an inability to increase gluconeogenesis. Liver triacylglycerides were elevated after acute exercise and circulating β-hydroxybutyrate was higher during postexercise refeeding in untrained KO mice. In contrast, exercise training prevented liver triacylglyceride accumulation in KO mice. This was accompanied by pronounced increases in indices of skeletal muscle mitochondrial oxidative metabolism in KO mice. Together, these results show that hepatic gluconeogenesis is dispensable for exercise training to reduce liver lipids. This may be due to responses in ketone body metabolism and/or metabolic adaptations in skeletal muscle to exercise.NEW & NOTEWORTHY Exercise training reduces hepatic steatosis partly through enhanced hepatic terminal oxidation. During acute exercise, hepatic gluconeogenesis is elevated to match the heightened rate of muscle glucose uptake and maintain glucose homeostasis. It has been postulated that the hepatic energetic stress induced by elevating gluconeogenesis during acute exercise is a key stimulus underlying the beneficial metabolic responses to exercise training. This study shows that hepatic gluconeogenesis is not necessary for exercise training to lower liver lipids.
Collapse
Affiliation(s)
- Ferrol I Rome
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Gregory L Shobert
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - William C Voigt
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - David B Stagg
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Shawn C Burgess
- Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Curtis C Hughey
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
25
|
Fuller KNZ, McCoin CS, Stierwalt H, Allen J, Gandhi S, Perry CGR, Jambal P, Shankar K, Thyfault JP. Oral combined contraceptives induce liver mitochondrial reactive oxygen species and whole-body metabolic adaptations in female mice. J Physiol 2022; 600:5215-5245. [PMID: 36326014 DOI: 10.1113/jp283733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Compared to age-matched men, pre-menopausal women show greater resilience against cardiovascular disease (CVD), hepatic steatosis, diabetes and obesity - findings that are widely attributed to oestrogen. However, meta-analysis data suggest that current use of oral combined contraceptives (OC) is a risk factor for myocardial infarction, and OC use further compounds with metabolic disease risk factors to increase CVD susceptibility. While mitochondrial function in tissues such as the liver and skeletal muscle is an emerging mechanism by which oestrogen may confer its protection, effects of OC use on mitochondria and metabolism in the context of disease risk remain unexplored. To answer this question, female C57Bl/6J mice were fed a high fat diet and treated with vehicle or OCs for 3, 12 or 20 weeks (n = 6 to 12 per group) at a dose and ratio that mimic the human condition of cycle cessation in the low oestrogen, high progesterone stage. Liver and skeletal muscle mitochondrial function (respiratory capacity, H2 O2 , coupling) was measured along with clinical outcomes of cardiometabolic disease such as obesity, glucose tolerance, hepatic steatosis and aortic atherosclerosis. The main findings indicate that regardless of treatment duration, OCs robustly increase hepatic mitochondrial H2 O2 levels, likely due to diminished antioxidant capacity, but have no impact on muscle mitochondrial H2 O2 . Furthermore, OC-treated mice had lower adiposity and hepatic triglyceride content compared to control mice despite reduced wheel running, spontaneous physical activity and total energy expenditure. Together, these studies describe tissue-specific effects of OC use on mitochondria as well as variable impacts on markers of metabolic disease susceptibility. KEY POINTS: Oestrogen loss in women increases risk for cardiometabolic diseases, a link that has been partially attributed to negative impacts on mitochondria and energy metabolism. To study the effect of oral combined contraceptives (OCs) on hepatic and skeletal muscle mitochondria and whole-body energy metabolism, we used an animal model of OCs which mimics the human condition of cessation of hormonal cycling in the low oestrogen, high progesterone state. OC-treated mice have increased hepatic mitochondrial oxidative stress and decreased physical activity and energy expenditure, despite displaying lower adiposity and liver fat at this time point. These pre-clinical data reveal tissue-specific effects of OCs that likely underlie the clinical findings of increased cardiometabolic disease in women who use OCs compared to non-users, when matched for obesity.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Colin S McCoin
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA.,Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.,University of Kansas Diabetes Institute, Kansas City, KS, USA.,Kansas Center for Metabolism and Obesity Research, Kansas City, KS, USA
| | - Harrison Stierwalt
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Julie Allen
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Center, York University, Toronto, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Science, Muscle Health Research Center, York University, Toronto, Canada
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine Anschutz Medical Campus, Aurora, CO, USA
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA.,Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA.,University of Kansas Diabetes Institute, Kansas City, KS, USA.,Kansas Center for Metabolism and Obesity Research, Kansas City, KS, USA.,Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
26
|
McCoin CS, Franczak E, Washburn MP, Sardiu ME, Thyfault JP. Acute exercise dynamically modulates the hepatic mitochondrial proteome. Mol Omics 2022; 18:840-852. [PMID: 35929479 PMCID: PMC9633379 DOI: 10.1039/d2mo00143h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exercise powerfully increases energy metabolism and substrate flux in tissues, a process reliant on dramatic changes in mitochondrial energetics. Liver mitochondria play a multi-factorial role during exercise to fuel hepatic glucose output. We previously showed acute exercise activates hepatic mitophagy, a pathway to recycle low-functioning/damaged mitochondria, however little is known how individual bouts of exercise alters the hepatic mitochondrial proteome. Here we leveraged proteomics to examine changes in isolated hepatic mitochondria both immediately after and 2 hours post an acute, 1 hour bout of treadmill exercise in female mice. Further, we utilized leupeptin, a lysosomal inhibitor, to capture and measure exercise-induced changes in mitochondrial proteins that would have been unmeasured due to their targeting for lysosomal degradation. Proteomic analysis of enriched hepatic mitochondria identified 3241 total proteins. Functional enrichment analysis revealed robust enrichment for proteins critical to the mitochondria including metabolic pathways, tricarboxylic acid cycle, and electron transport system. Compared to the sedentary condition, exercise elevated processes regulating lipid localization, Il-5 signaling, and protein phosphorylation in isolated mitochondria. t-SNE analysis identified 4 unique expressional clusters driven by time-dependent changes in protein expression. Isolation of proteins significantly altered with exercise from each cluster revealed influences of leupeptin and exercise both independently and cooperatively modulating mitochondrial protein expressional profiles. Overall, we provide evidence that acute exercise rapidly modulates changes in the proteins/pathways of isolated hepatic mitochondria that include fatty acid metabolism/storage, post-translational protein modification, inflammation, and oxidative stress. In conclusion, the hepatic mitochondrial proteome undergoes extensive remodeling with a bout of exercise.
Collapse
Affiliation(s)
- Colin S McCoin
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, 64128, USA
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity Research, Kansas City, MO, 64128, USA
| | - Edziu Franczak
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Michael P Washburn
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mihaela E Sardiu
- Department of Biostatistics and Data Science, The University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - John P Thyfault
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, 64128, USA
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity Research, Kansas City, MO, 64128, USA
- Department of Internal Medicine-Division of Endocrinology and Metabolism, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Kansas City Veterans Affairs Medical Center, Kansas City, MO, 64128, USA
| |
Collapse
|
27
|
Cui B, Zhu Y, Zhang X, He K, Shi Y, Yu J, Zhou W, Zhu Y, Yan H. Association of Physical Activity with Retinal Thickness and Vascular Structure in Elderly Chinese Population. Ophthalmic Res 2022; 66:281-292. [PMID: 36252557 DOI: 10.1159/000527448] [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: 08/19/2022] [Accepted: 09/30/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION We aimed to assess the association of physical activity (PA) with retinal thickness and vascular structure in an elderly Chinese population. METHODS This study enrolled 220 retirees aged 50 years and above from Tianjin University of Sport, China. PA data gathered through the International Physical Activity Questionnaires were computed for metabolic equivalent of task-minutes per week. All participants underwent full ophthalmic examination including optical coherence tomography angiography. Multiple linear regression analyses were used to evaluate the association of PA with retinal thickness and vascular structure. RESULTS A high amount of total PA was associated with smaller size of foveal avascular zone (FAZ) (high vs. low: area, β = -0.04; circumference β = -0.14) and greater central vessel density (VD) (β = 1.04) and perfusion density (PD) (β = 0.02). For domain-specific PA, participants with high and moderate recreational PA levels were found to have greater central VDs and PDs. Nonleisure activities demonstrated negative relationship with FAZ perimeter ratio (moderate vs. low: β = -0.08; high vs. low: β = -0.13) and, counter-intuitively, negative relation with VDs measured at the inner (high vs. low: β = -0.98) and outer retinal layers (high vs. low: β = -0.38). Additionally, higher levels of total PA were related to increased macular fovea thickness and average thickness of retinal nerve fiber layer. CONCLUSIONS High PA level, particularly recreational PA, was associated with smaller FAZ and greater VD and PD in senior adults. Nonleisure-time PA might not confer the same health benefits as leisure-time PA. Our data support a possible protective role of recreational PA in the retina against microvascular changes.
Collapse
Affiliation(s)
- Bohao Cui
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaodan Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Kai He
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Shi
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jinguo Yu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Zhou
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yun Zhu
- Department of Epidemiology and Biostatistics, School of Public Health Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
28
|
Xiao J, Li X, Zhou Z, Guan S, Zhuo L, Gao B. Development of an in vitro insulin resistance dissociated model of hepatic steatosis by co-culture system. Biosci Trends 2022; 16:257-266. [PMID: 35965099 DOI: 10.5582/bst.2022.01242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The evidence shows that there is an associated relationship between hepatosteatosis and insulin resistance. While some existing genetic induction animal and patient models challenge this relationship, indicating that hepatosteatosis is dissociated from insulin resistance. However, the molecular mechanisms of this dissociation remain poorly understood due to a lack of available, reliable, and simplistic setup models. Currently, we used primary rat hepatocytes (rHPCs), co-cultured with rat hepatic stellate cells (HSC-T6) or human foreskin fibroblast cells (HFF-1) in stimulation with high insulin and glucose, to develop a model of steatosis charactered as dissociated lipid accumulation from insulin resistance. Oil-Red staining significantly showed intracellular lipid accumulated in the developed model. Gene expression of sterol regulatory element-binding protein 1c (SREBP1c) and elongase of very-long-chain fatty acids 6 (ELOVL6), key genes responsible for lipogenesis, were detected and obviously increased in this model. Inversely, the insulin resistance related genes expression included phosphoenolpyruvate carboxykinase 1 (PCK1), pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4), and glucose-6-phosphatase (G6pase) were decreased, suggesting a dissociation relationship between steatosis and insulin resistance in the developed model. As well, the drug metabolism of this developed model was investigated and showed up-regulation of cytochrome P450 3A (CYP3A) and down-regulation of cytochrome P450 2E1 (CYP2E1) and cytochrome P450 1A2 (CYP1A2). Taken together, those results demonstrate that the in vitro model of dissociated steatosis from insulin resistance was successfully created by our co-cultured cells in high insulin and glucose medium, which will be a potential model for investigating the mechanism of insulin resistance dissociated steatosis, and discovering a novel drug for its treatment.
Collapse
Affiliation(s)
- Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Xiang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zongbao Zhou
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Shuwen Guan
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Lingjian Zhuo
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| |
Collapse
|
29
|
Stierwalt HD, Morris EM, Maurer A, Apte U, Phillips K, Li T, Meers GME, Koch LG, Britton SL, Graf G, Rector RS, Mercer K, Shankar K, Thyfault JP. Rats with high aerobic capacity display enhanced transcriptional adaptability and upregulation of bile acid metabolism in response to an acute high-fat diet. Physiol Rep 2022; 10:e15405. [PMID: 35923133 PMCID: PMC9350427 DOI: 10.14814/phy2.15405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 06/09/2023] Open
Abstract
Rats selectively bred for the high intrinsic aerobic capacity runner (HCR) or low aerobic capacity runner (LCR) show pronounced differences in susceptibility for high-fat/high sucrose (HFHS) diet-induced hepatic steatosis and insulin resistance, replicating the protective effect of high aerobic capacity in humans. We have previously shown multiple systemic differences in energy and substrate metabolism that impacts steatosis between HCR and LCR rats. This study aimed to investigate hepatic-specific mechanisms of action via changes in gene transcription. Livers of HCR rats had a greater number of genes that significantly changed in response to 3-day HFHS compared with LCR rats (171 vs. 75 genes: >1.5-fold, p < 0.05). HCR and LCR rats displayed numerous baseline differences in gene expression while on a low-fat control diet (CON). A 3-day HFHS diet resulted in greater expression of genes involved in the conversion of excess acetyl-CoA to cholesterol and bile acid (BA) synthesis compared with the CON diet in HCR, but not LCR rats. These results were associated with higher fecal BA loss and lower serum BA concentrations in HCR rats. Exercise studies in rats and mice also revealed higher hepatic expression of cholesterol and BA synthesis genes. Overall, these results suggest that high aerobic capacity and exercise are associated with upregulated BA synthesis paired with greater fecal excretion of cholesterol and BA, an effect that may play a role in protection against hepatic steatosis in rodents.
Collapse
Affiliation(s)
- Harrison D. Stierwalt
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| | - E. Matthew Morris
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Adrianna Maurer
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and TherapeuticsUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | | | - Tiangang Li
- Department of PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Grace M. E. Meers
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
| | - Lauren G. Koch
- Physiology and PharmacologyThe University of ToledoToledoOhioUSA
| | | | - Greg Graf
- Department of Pharmaceutical SciencesSaha Cardiovascular Research Center, University of KentuckyLexingtonKentuckyUSA
| | - R. Scott Rector
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
- Research ServiceHarry S Truman Memorial VA HospitalColumbiaMissouriUSA
| | - Kelly Mercer
- Arkansas Children's Nutrition CenterUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Kartik Shankar
- Section of Nutrition, Department of PediatricsUniversity of Colorado School of Medicine Anschutz Medical CampusAuroraColoradoUSA
| | - John P. Thyfault
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| |
Collapse
|
30
|
CT-based visual grading system for assessment of hepatic steatosis: diagnostic performance and interobserver agreement. Hepatol Int 2022; 16:1075-1084. [PMID: 35789473 DOI: 10.1007/s12072-022-10373-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/30/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Hepatic steatosis (HS) can be comprehensively assessed by visually comparing the hepatic and vessel attenuation on unenhanced computed tomography (CT). We aimed to evaluate the reliability and reproducibility of a CT-based visual grading system (VGS) for comprehensive assessment of HS. METHODS In this retrospective study, a four-point VGS based on the visual comparison of liver and hepatic vessels was validated by six reviewers with diverse clinical experience using the unenhanced CT images of 717 potential liver donors. The diagnostic performance of VGS and quantitative indices (difference and ratio of the hepatic and splenic attenuation) to diagnose HS were evaluated using multi-reader multi-case receiver operating characteristics (ROC) analysis (reference: pathology). The interobserver agreement was assessed using Fleiss κ statistics. RESULTS Using the VGS, all six reviewers showed areas under the ROC curves (AUROCs) higher than 0.9 for diagnosing total steatosis (TS) ≥ 30%, macrovesicular steatosis (MaS) ≥ 30%, and MaS ≥ 10%. No difference was noted between the AUROCs of the VGS and quantitative indices (p ≥ 0.1). The reviewers showed substantial agreement (Fleiss κ, 0.61). Most discrepancies occurred between the two lowest grades of VGS (81.5%; 233/283), in which most subjects (97.0%; 226/233) had a MaS < 10%. The average-reader sensitivity and specificity of the VGS were 0.80 and 0.94 to detect TS ≥ 30% and 0.93 and 0.81 to detect MaS ≥ 10%. CONCLUSION VGS was reliable and reproducible in assessing HS. It may be useful as a non-invasive and simple tool for comprehensive HS assessment.
Collapse
|
31
|
NAFLD: Mechanisms, Treatments, and Biomarkers. Biomolecules 2022; 12:biom12060824. [PMID: 35740949 PMCID: PMC9221336 DOI: 10.3390/biom12060824] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently renamed metabolic-associated fatty liver disease (MAFLD), is one of the most common causes of liver diseases worldwide. NAFLD is growing in parallel with the obesity epidemic. No pharmacological treatment is available to treat NAFLD, specifically. The reason might be that NAFLD is a multi-factorial disease with an incomplete understanding of the mechanisms involved, an absence of accurate and inexpensive imaging tools, and lack of adequate non-invasive biomarkers. NAFLD consists of the accumulation of excess lipids in the liver, causing lipotoxicity that might progress to metabolic-associated steatohepatitis (NASH), liver fibrosis, and hepatocellular carcinoma. The mechanisms for the pathogenesis of NAFLD, current interventions in the management of the disease, and the role of sirtuins as potential targets for treatment are discussed here. In addition, the current diagnostic tools, and the role of non-coding RNAs as emerging diagnostic biomarkers are summarized. The availability of non-invasive biomarkers, and accurate and inexpensive non-invasive diagnosis tools are crucial in the detection of the early signs in the progression of NAFLD. This will expedite clinical trials and the validation of the emerging therapeutic treatments.
Collapse
|
32
|
Antunes GC, Lima RDD, Vieira RFL, Macêdo APA, Muñoz VR, Zambalde EP, Romeiro CF, Simabuco FM, Prada PO, da Silva ASR, Ropelle ER, Cintra DE, Pauli JR. RESISTANCE EXERCISE ATTENUATES IKKε PHOSPHORYLATION AND HEPATIC FAT ACCUMULATION OF OBESE MICE. Clin Exp Pharmacol Physiol 2022; 49:1072-1081. [PMID: 35690890 DOI: 10.1111/1440-1681.13687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/12/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Obesity is associated with low-grade inflammation and disturbances in hepatic metabolism. This study aimed to investigate the effects of resistance exercise on inflammatory signaling related to IKKepsilon protein (IKKɛ) and on hepatic fat accumulation in obese mice. Male Swiss mice were distributed into three groups: control (CTL) fed with standard chow; obese (OB) mice induced by a high-fat diet (HFD); obese exercised (OB+RE) mice fed with HFD and submitted to a resistance exercise training. The resistance exercise training protocol consisted of 20 sets/3 ladder climbs for eight weeks, three times/week on alternate days. The training overload was equivalent to 70% of the maximum load supported by the rodent. Assays were performed to evaluate weight gain, hepatic fat content, fasting glucose, insulin sensitivity, IKKɛ phosphorylation, and proteins related to insulin signaling and lipogenesis in the liver. Mice that received the high-fat diet showed greater adiposity, impaired insulin sensitivity, increased fasting glucose, and increased hepatic fat accumulation. These results were accompanied by an increase in IKKɛ phosphorylation and lipogenesis-related proteins such as cluster of differentiation 36 (CD36) and fatty acid synthase (FAS) in the liver of obese mice. In contrast, exercised mice showed lower body weight and adiposity evolution throughout the experiment. In addition, resistance exercise suppressed the effects of the high-fat diet by reducing IKKɛ phosphorylation and hepatic fat content. In conclusion, resistance exercise training improves hepatic fat metabolism and glycemic homeostasis, which are, at least in part, linked to the antiinflammatory effect of reduced IKKɛ phosphorylation in the liver of obese mice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Gabriel Calheiros Antunes
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Robson Damasceno de Lima
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana Paula Azevêdo Macêdo
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Erika Pereira Zambalde
- Multidisciplinary Laboratory of Food and Health, State University of Campinas, Faculty of Applied Sciences, Limeira, São Paulo, Brazil
| | - Caio Felipe Romeiro
- Multidisciplinary Laboratory of Food and Health, State University of Campinas, Faculty of Applied Sciences, Limeira, São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health, State University of Campinas, Faculty of Applied Sciences, Limeira, São Paulo, Brazil
| | - Patricia Oliveira Prada
- Laboratory of Molecular Research in Obesity (Labimo), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, and Postgraduate Program in Physical Education and Sport, School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Dennys Esper Cintra
- OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
33
|
Nardo WD, Miotto PM, Bayliss J, Nie S, Keenan SN, Montgomery MK, Watt MJ. Proteomic analysis reveals exercise training induced remodelling of hepatokine secretion and uncovers syndecan-4 as a regulator of hepatic lipid metabolism. Mol Metab 2022; 60:101491. [PMID: 35381388 PMCID: PMC9034320 DOI: 10.1016/j.molmet.2022.101491] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/04/2022] Open
Abstract
Objective Non-alcoholic fatty liver disease (NAFLD) is linked to impaired lipid metabolism and systemic insulin resistance, which is partly mediated by altered secretion of liver proteins known as hepatokines. Regular physical activity can resolve NAFLD and improve its metabolic comorbidities, however, the effects of exercise training on hepatokine secretion and the metabolic impact of exercise-regulated hepatokines in NAFLD remain unresolved. Herein, we examined the effect of endurance exercise training on hepatocyte secreted proteins with the aim of identifying proteins that regulate metabolism and reduce NAFLD severity. Methods C57BL/6 mice were fed a high-fat diet for six weeks to induce NAFLD. Mice were exercise trained for a further six weeks, while the control group remained sedentary. Hepatocytes were isolated two days after the last exercise bout, and intracellular and secreted proteins were detected using label-free mass spectrometry. Hepatocyte secreted factors were applied to skeletal muscle and liver ex vivo and insulin action and fatty acid metabolism were assessed. Syndecan-4 (SDC4), identified as an exercise-responsive hepatokine, was overexpressed in the livers of mice using adeno-associated virus. Whole-body energy homeostasis was assessed by indirect calorimetry and skeletal muscle and liver metabolism was assessed using radiometric techniques. Results Proteomics analysis detected 2657 intracellular and 1593 secreted proteins from mouse hepatocytes. Exercise training remodelled the hepatocyte proteome, with differences in 137 intracellular and 35 secreted proteins. Bioinformatic analysis of hepatocyte secreted proteins revealed enrichment of tumour suppressive proteins and proteins involved in lipid metabolism and mitochondrial function, and suppression of oncogenes and regulators of oxidative stress. Hepatocyte secreted factors from exercise trained mice improved insulin action in skeletal muscle and increased hepatic fatty acid oxidation. Hepatocyte-specific overexpression of SDC4 reduced hepatic steatosis, which was associated with reduced hepatic fatty acid uptake, and blunted pro-inflammatory and pro-fibrotic gene expression. Treating hepatocytes with recombinant ectodomain of SDC4 (secreted form) recapitulated these effects with reduced fatty acid uptake, lipid storage and lipid droplet accumulation. Conclusions Remodelling of hepatokine secretion is an adaptation to regular exercise training that induces changes in metabolism in the liver and skeletal muscle. SDC4 is a novel exercise-responsive hepatokine that decreases fatty acid uptake and reduces steatosis in the liver. By understanding the proteomic changes in hepatocytes with exercise, these findings have potential for the discovery of new therapeutic targets for NAFLD. Exercise training remodels hepatokine secretion. Exercise regulated secreted factors improve insulin action in skeletal muscle. Syndecan-4 (SDC4) is a novel exercise-induced hepatokine. SDC4 reduces hepatic fatty acid uptake and hepatic steatosis.
Collapse
|
34
|
Moore MP, Wieschhaus NE, Alessi IG. The impact of time-restricted feeding in combination with aerobic exercise on weight gain and markers of hepatic metabolism. J Physiol 2022; 600:2277-2278. [PMID: 35362100 DOI: 10.1113/jp283020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 03/29/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Mary P Moore
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Nicole E Wieschhaus
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211
| | | |
Collapse
|
35
|
CTRP7 Is a Biomarker Related to Insulin Resistance and Oxidative Stress: Cross-Sectional and Intervention Studies In Vivo and In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6877609. [PMID: 35368863 PMCID: PMC8967592 DOI: 10.1155/2022/6877609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/17/2022] [Indexed: 12/17/2022]
Abstract
Objective CTRP7 is a cytokine that is known to be associated with obesity. However, its relationship with insulin resistance (IR) and metabolic diseases remains unknown. The aim of this study is to investigate the relationship between CTRP7 and IR under in vivo and in vitro conditions. Methods CTRP7 expression in mice and hepatocytes was determined using RT-qPCR and western blotting. Circulating CTRP7 concentrations were measured with an ELISA kit. EHC, OGTT, lipid-infusion, physical activity, and cold-stimulation experiments were performed in humans and mice. SOD, GSH, and MDA were measured by commercial kits. ROS levels were detected using dichlorofluorescein diacetate. Results The expression levels of CTRP7 protein in the liver and fat of ob/ob and db/db mice were higher than that of WT mice. Individuals with IGT, T2DM, and obesity had higher circulating CTRP7 levels. CTRP7 levels were associated with HOMA-IR, obesity, and other metabolic parameters. During OGTT, serum CTRP7 levels gradually decreased, while CTRP7 levels significantly increased during EHC in response to hyperinsulinemia in healthy individuals without IR. In addition, lipid infusion-induced IR further increased serum CTRP7 levels in healthy adults. Physical activity increased serum CTRP7 levels in healthy individuals and CTRP7 protein expression in iWAT and skeletal muscle in mice. Under in vitro conditions, the expression of the CTRP7 protein was inhibited in a glucose concentration-dependent manner but was promoted by FFAs and insulin stimulation in hepatocytes. Furthermore, CTRP7 overexpression facilitated oxidative stress and suppressed the phosphorylation of insulin signaling molecules in hepatocytes. Conclusions Our evidence shows that CTRP7 could be a useful biomarker and potential treatment target in IR and metabolic disorders.
Collapse
|
36
|
McCoin CS, Franczak E, Deng F, Pei D, Ding WX, Thyfault JP. Acute exercise rapidly activates hepatic mitophagic flux. J Appl Physiol (1985) 2022; 132:862-873. [PMID: 35142562 PMCID: PMC8934677 DOI: 10.1152/japplphysiol.00704.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/25/2022] [Accepted: 02/02/2022] [Indexed: 01/18/2023] Open
Abstract
Exercise is critical for improving metabolic health and putatively maintains or enhances mitochondrial quality control in metabolic tissues. Although previous work has shown that exercise elicits hepatic mitochondrial biogenesis, it is unknown if acute exercise activates hepatic mitophagy, the selective degradation of damaged or low-functioning mitochondria. We tested if an acute bout of treadmill running increased hepatic mitophagic flux both right after and 2-h postexercise in 15- to 24-wk-old C57BL/6J female mice. Acute exercise did not significantly increase markers of autophagic flux, however, mitophagic flux was activated 2-h post-treadmill running as measured by accumulation of both LC3-II and p62 in isolated mitochondria in the presence of leupeptin, an inhibitor of autophagosome degradation. Furthermore, mitochondrial-associated ubiquitin, which recruits the autophagy receptor protein p62, was also significantly increased at 2 h. Further examination via Western blot and proteomics analysis revealed that acute exercise elicits a time-dependent, dynamic activation of mitophagy pathways. Moreover, the results suggest that exercise-induced hepatic mitophagy is likely mediated by both polyubiquitination and receptor-mediated signaling pathways. Overall, we provide evidence that acute exercise activates hepatic mitophagic flux while also revealing specific receptor-mediated proteins by which exercise maintains mitochondrial quality control in the liver.NEW & NOTEWORTHY This study provides evidence that acute exercise activates hepatic mitophagic flux and mitochondrial polyubiquitination while additionally revealing specific receptor-mediated proteins by which exercise maintains mitochondrial quality control in the liver.
Collapse
Affiliation(s)
- Colin S McCoin
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Kansas City, Kansas City, Missouri
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - Edziu Franczak
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
| | - Fengyan Deng
- Stowers Institute for Medical Research, Kansas City, Missouri
| | - Dong Pei
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Missouri
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Children's Mercy Kansas City, Kansas City, Missouri
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| |
Collapse
|
37
|
Kim J, Mondaca-Ruff D, Singh S, Wang Y. SIRT1 and Autophagy: Implications in Endocrine Disorders. Front Endocrinol (Lausanne) 2022; 13:930919. [PMID: 35909524 PMCID: PMC9331929 DOI: 10.3389/fendo.2022.930919] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/20/2022] [Indexed: 11/26/2022] Open
Abstract
Autophagy is a cellular process involved in the selective degradation and recycling of dysfunctional intracellular components. It plays a crucial role in maintaining cellular homeostasis and survival by removing damaged and harmful proteins, lipids, and organelles. SIRT1, an NAD+-dependent multifunctional enzyme, is a key regulator of the autophagy process. Through its deacetylase activity, SIRT1 participates in the regulation of different steps of autophagy, from initiation to degradation. The levels and function of SIRT1 are also regulated by the autophagy process. Dysregulation in SIRT1-mediated autophagy hinders the proper functioning of the endocrine system, contributing to the onset and progression of endocrine disorders. This review provides an overview of the crosstalk between SIRT1 and autophagy and their implications in obesity, type-2 diabetes mellitus, diabetic cardiomyopathy, and hepatic steatosis.
Collapse
|
38
|
Non-alcoholic fatty liver disease in obese children and adolescents: a role for nutrition? Eur J Clin Nutr 2022; 76:28-39. [PMID: 34006994 DOI: 10.1038/s41430-021-00928-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/06/2021] [Accepted: 04/16/2021] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease in children, paralleling the increasing prevalence of obesity worldwide. The pathogenesis of paediatric NAFLD is not fully understood, but it is known that obesity, nutrition, lifestyle variables, genetic and epigenetic factors may be causally involved in the development of this common metabolic liver disease. In particular, obesity and nutrition are among the strongest risk factors for paediatric NAFLD, which may exert their adverse hepatic effects already before birth. Excess energy intake induces hypertrophy and hyperplasia of adipose tissue with subsequent development of systemic insulin resistance, which is another important risk factor for NAFLD. Diet composition and in particular simple carbohydrate intake (especially high fructose intake) may promote the development of NAFLD, whereas non-digestible carbohydrates (dietary fiber), by affecting gut microbiota, may favour the integrity of gut wall and reduce inflammation, opposing this process. Saturated fat intake may also promote NAFLD development, whereas unsaturated fat intake has some beneficial effects. Protein intake does not seem to affect the development of NAFLD, but further investigation is needed. In conclusion, lifestyle modifications to induce weight loss, through diet and physical activity, remain the mainstay of treatment for paediatric NAFLD. The use of dietary supplements, such as omega-3 fatty acids and probiotics, needs further study before recommendation.
Collapse
|
39
|
Interactions between insulin and exercise. Biochem J 2021; 478:3827-3846. [PMID: 34751700 DOI: 10.1042/bcj20210185] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023]
Abstract
The interaction between insulin and exercise is an example of balancing and modifying the effects of two opposing metabolic regulatory forces under varying conditions. While insulin is secreted after food intake and is the primary hormone increasing glucose storage as glycogen and fatty acid storage as triglycerides, exercise is a condition where fuel stores need to be mobilized and oxidized. Thus, during physical activity the fuel storage effects of insulin need to be suppressed. This is done primarily by inhibiting insulin secretion during exercise as well as activating local and systemic fuel mobilizing processes. In contrast, following exercise there is a need for refilling the fuel depots mobilized during exercise, particularly the glycogen stores in muscle. This process is facilitated by an increase in insulin sensitivity of the muscles previously engaged in physical activity which directs glucose to glycogen resynthesis. In physically trained individuals, insulin sensitivity is also higher than in untrained individuals due to adaptations in the vasculature, skeletal muscle and adipose tissue. In this paper, we review the interactions between insulin and exercise during and after exercise, as well as the effects of regular exercise training on insulin action.
Collapse
|
40
|
Hoene M, Kappler L, Kollipara L, Hu C, Irmler M, Bleher D, Hoffmann C, Beckers J, Hrabě de Angelis M, Häring HU, Birkenfeld AL, Peter A, Sickmann A, Xu G, Lehmann R, Weigert C. Exercise prevents fatty liver by modifying the compensatory response of mitochondrial metabolism to excess substrate availability. Mol Metab 2021; 54:101359. [PMID: 34695608 PMCID: PMC8671118 DOI: 10.1016/j.molmet.2021.101359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023] Open
Abstract
Objective Liver mitochondria adapt to high-calorie intake. We investigated how exercise alters the early compensatory response of mitochondria, thus preventing fatty liver disease as a long-term consequence of overnutrition. Methods We compared the effects of a steatogenic high-energy diet (HED) for six weeks on mitochondrial metabolism of sedentary and treadmill-trained C57BL/6N mice. We applied multi-OMICs analyses to study the alterations in the proteome, transcriptome, and lipids in isolated mitochondria of liver and skeletal muscle as well as in whole tissue and examined the functional consequences by high-resolution respirometry. Results HED increased the respiratory capacity of isolated liver mitochondria, both in sedentary and in trained mice. However, proteomics analysis of the mitochondria and transcriptomics indicated that training modified the adaptation of the hepatic metabolism to HED on the level of respiratory complex I, glucose oxidation, pyruvate and acetyl-CoA metabolism, and lipogenesis. Training also counteracted the HED-induced glucose intolerance, the increase in fasting insulin, and in liver fat by lowering diacylglycerol species and c-Jun N-terminal kinase (JNK) phosphorylation in the livers of trained HED-fed mice, two mechanisms that can reverse hepatic insulin resistance. In skeletal muscle, the combination of HED and training improved the oxidative capacity to a greater extent than training alone by increasing respiration of isolated mitochondria and total mitochondrial protein content. Conclusion We provide a comprehensive insight into the early adaptations of mitochondria in the liver and skeletal muscle to HED and endurance training. Our results suggest that exercise disconnects the HED-induced increase in mitochondrial substrate oxidation from pyruvate and acetyl-CoA-driven lipid synthesis. This could contribute to the prevention of deleterious long-term effects of high fat and sugar intake on hepatic mitochondrial function and insulin sensitivity. High-energy diet promotes mitochondrial respiration in liver independent of training. High-energy diet combined with training disconnects substrate oxidation from lipid synthesis. High-energy diet combined with training reduces complex I formation in the liver. Trained skeletal muscle unburdens the liver from substrate overload. Comprehensive resource of mitochondrial adaptations to high-energy diet and training.
Collapse
Affiliation(s)
- Miriam Hoene
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Lisa Kappler
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Martin Irmler
- Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764, Neuherberg, Germany
| | - Daniel Bleher
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Christoph Hoffmann
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Johannes Beckers
- Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764, Neuherberg, Germany; Technische Universität München, Chair of Experimental Genetics, 85354, Freising, Germany; German Center for Diabetes Research (DZD), Germany
| | - Martin Hrabě de Angelis
- Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), 85764, Neuherberg, Germany; Technische Universität München, Chair of Experimental Genetics, 85354, Freising, Germany; German Center for Diabetes Research (DZD), Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen, Tuebingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen, Tuebingen, Germany; Department of Internal Medicine IV, University Hospital Tuebingen, Tuebingen, Germany
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany; German Center for Diabetes Research (DZD), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen, Tuebingen, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Rainer Lehmann
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany; German Center for Diabetes Research (DZD), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen, Tuebingen, Germany
| | - Cora Weigert
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tuebingen, Tuebingen, Germany; German Center for Diabetes Research (DZD), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
41
|
Cunningham RP, Porat-Shliom N. Liver Zonation - Revisiting Old Questions With New Technologies. Front Physiol 2021; 12:732929. [PMID: 34566696 PMCID: PMC8458816 DOI: 10.3389/fphys.2021.732929] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the ever-increasing prevalence of non-alcoholic fatty liver disease (NAFLD), the etiology and pathogenesis remain poorly understood. This is due, in part, to the liver's complex physiology and architecture. The liver maintains glucose and lipid homeostasis by coordinating numerous metabolic processes with great efficiency. This is made possible by the spatial compartmentalization of metabolic pathways a phenomenon known as liver zonation. Despite the importance of zonation to normal liver function, it is unresolved if and how perturbations to liver zonation can drive hepatic pathophysiology and NAFLD development. While hepatocyte heterogeneity has been identified over a century ago, its examination had been severely hindered due to technological limitations. Recent advances in single cell analysis and imaging technologies now permit further characterization of cells across the liver lobule. This review summarizes the advances in examining liver zonation and elucidating its regulatory role in liver physiology and pathology. Understanding the spatial organization of metabolism is vital to further our knowledge of liver disease and to provide targeted therapeutic avenues.
Collapse
Affiliation(s)
- Rory P Cunningham
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Natalie Porat-Shliom
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
42
|
Babu AF, Csader S, Lok J, Gómez-Gallego C, Hanhineva K, El-Nezami H, Schwab U. Positive Effects of Exercise Intervention without Weight Loss and Dietary Changes in NAFLD-Related Clinical Parameters: A Systematic Review and Meta-Analysis. Nutrients 2021; 13:nu13093135. [PMID: 34579012 PMCID: PMC8466505 DOI: 10.3390/nu13093135] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/05/2021] [Accepted: 09/05/2021] [Indexed: 12/13/2022] Open
Abstract
One of the focuses of non-alcoholic fatty liver disease (NAFLD) treatment is exercise. Randomized controlled trials investigating the effects of exercise without dietary changes on NAFLD-related clinical parameters (liver parameters, lipid metabolism, glucose metabolism, gut microbiota, and metabolites) were screened using the PubMed, Scopus, Web of Science, and Cochrane databases on 13 February 2020. Meta-analyses were performed on 10 studies with 316 individuals who had NAFLD across three exercise regimens: aerobic exercise, resistance training, and a combination of both. No studies investigating the role of gut microbiota and exercise in NAFLD were found. A quality assessment via the (RoB)2 tool was conducted and potential publication bias, statistical outliers, and influential cases were identified. Overall, exercise without significant weight loss significantly reduced the intrahepatic lipid (IHL) content (SMD: −0.76, 95% CI: −1.04, −0.48) and concentrations of alanine aminotransaminase (ALT) (SMD: −0.52, 95% CI: −0.90, −0.14), aspartate aminotransaminase (AST) (SMD: −0.68, 95% CI: −1.21, −0.15), low-density lipoprotein cholesterol (SMD: −0.34, 95% CI: −0.66, −0.02), and triglycerides (TG) (SMD: −0.59, 95% CI: −1.16, −0.02). The concentrations of high-density lipoprotein cholesterol, total cholesterol (TC), fasting glucose, fasting insulin, and glycated hemoglobin were non-significantly altered. Aerobic exercise alone significantly reduced IHL, ALT, and AST; resistance training alone significantly reduced TC and TG; a combination of both exercise types significantly reduced IHL. To conclude, exercise overall likely had a beneficial effect on alleviating NAFLD without significant weight loss. The study was registered at PROSPERO: CRD42020221168 and funded by the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement no. 813781.
Collapse
Affiliation(s)
- Ambrin Farizah Babu
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
- Afekta Technologies Ltd., Yliopistonranta 1L, 70211 Kuopio, Finland
| | - Susanne Csader
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
| | - Johnson Lok
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
| | - Carlos Gómez-Gallego
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
| | - Kati Hanhineva
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
- Afekta Technologies Ltd., Yliopistonranta 1L, 70211 Kuopio, Finland
- Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, 20500 Turku, Finland
| | - Hani El-Nezami
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Ursula Schwab
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; (A.F.B.); (S.C.); (J.L.); (C.G.-G.); (K.H.); (H.E.-N.)
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70210 Kuopio, Finland
- Correspondence: ; Tel.: +358-403552791
| |
Collapse
|
43
|
Brown E, Hydes T, Hamid A, Cuthbertson DJ. Emerging and Established Therapeutic Approaches for Nonalcoholic Fatty Liver Disease. Clin Ther 2021; 43:1476-1504. [PMID: 34446271 DOI: 10.1016/j.clinthera.2021.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Nonalcoholic fatty liver disease (NAFLD), more recently referred to as metabolic-associated fatty liver disease, refers to a disease spectrum ranging from hepatic steatosis to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis, associated with hepatic complications (including liver fibrosis, cirrhosis, and hepatocellular carcinoma) and extrahepatic complications (particularly cardiometabolic complications, including type 2 diabetes and cardiovascular disease). Treatment options include lifestyle interventions (dietary modification and physical activity programs) and pharmacologic interventions. Treatment aims should be broad, with a hepatic focus (to improve/reverse hepatic inflammation, fibrosis, and steatohepatitis), ideally with additional extrahepatic effects affecting metabolic co-morbidities (eg, insulin resistance, glucose dysregulation, dyslipidemia), causing weight loss and affording cardiovascular protection. NASH and fibrosis represent the main histopathological features that warrant treatment to prevent disease progression. Despite a paucity of established treatments, the array of potential molecular targets, pathways, and potential treatments is continually evolving. The goal of this article was to provide a narrative review summarizing the emerging and more established therapeutic options considering the complex pathophysiology of NAFLD and the important long-term sequelae of this condition. METHODS The literature was reviewed by using PubMed, conference abstracts, and press releases from early-phase clinical studies to provide an overview of the evidence. FINDINGS As understanding of the pathophysiology of NASH/NAFLD evolves, drugs with different mechanisms of action, targeting different molecular targets and aberrant pathways that mediate hepatic steatosis, inflammation, and fibrosis, have been developed and are being tested in clinical trials. Pharmacologic therapies fall into 4 main categories according to the molecular targets/pathways they disrupt: (1) meta-bolic targets, targeting insulin resistance, hepatic de novo lipogenesis, or substrate utilization; (2) inflam-matory pathways, inhibiting inflammatory cell recruitment/signaling, reduce oxidative/endoplasmic reticulum stress or are antiapoptotic; (3) the liver-gut axis, which modulates bile acid enterohepatic circulation/signaling or alters gut microbiota; and (4) antifibrotic targets, targeting hepatic stellate cells, decrease collagen deposition or increase fibrinolysis. IMPLICATIONS Lifestyle modification must remain the cornerstone of treatment. Pharmacologic treatment is reserved for NASH or fibrosis, the presence of which requires histopathological confirmation. The disease complexity provides a strong rationale for combination therapies targeting multiple pathways simultaneously.
Collapse
Affiliation(s)
- Emily Brown
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom.
| | - T Hydes
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - A Hamid
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - D J Cuthbertson
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
44
|
Abstract
The reactions of the tricarboxylic acid (TCA) cycle allow the controlled combustion of fat and carbohydrate. In principle, TCA cycle intermediates are regenerated on every turn and can facilitate the oxidation of an infinite number of nutrient molecules. However, TCA cycle intermediates can be lost to cataplerotic pathways that provide precursors for biosynthesis, and they must be replaced by anaplerotic pathways that regenerate these intermediates. Together, anaplerosis and cataplerosis help regulate rates of biosynthesis by dictating precursor supply, and they play underappreciated roles in catabolism and cellular energy status. They facilitate recycling pathways and nitrogen trafficking necessary for catabolism, and they influence redox state and oxidative capacity by altering TCA cycle intermediate concentrations. These functions vary widely by tissue and play emerging roles in disease. This article reviews the roles of anaplerosis and cataplerosis in various tissues and discusses how they alter carbon transitions, and highlights their contribution to mechanisms of disease. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Melissa Inigo
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| | - Stanisław Deja
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; .,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Shawn C Burgess
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; .,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
45
|
Fuller KNZ, McCoin CS, Von Schulze AT, Houchen CJ, Choi MA, Thyfault JP. Estradiol treatment or modest exercise improves hepatic health and mitochondrial outcomes in female mice following ovariectomy. Am J Physiol Endocrinol Metab 2021; 320:E1020-E1031. [PMID: 33870713 PMCID: PMC8285602 DOI: 10.1152/ajpendo.00013.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/16/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
We recently reported that compared with males, female mice have increased hepatic mitochondrial respiratory capacity and are protected against high-fat diet-induced steatosis. Here, we sought to determine the role of estrogen in hepatic mitochondrial function, steatosis, and bile acid metabolism in female mice and investigate potential benefits of exercise in the absence or presence of estrogen via ovariectomy (OVX). Female C57BL mice (n = 6 per group) were randomly assigned to sham surgery (sham), ovariectomy (OVX), or OVX plus estradiol replacement therapy (OVX + Est). Half of the mice in each treatment group were sedentary (SED) or had access to voluntary wheel running (VWR). All mice were fed a high-fat diet (HFD) and were housed at thermoneutral temperatures. We assessed isolated hepatic mitochondrial respiratory capacity using the Oroboros O2k with both pyruvate and palmitoylcarnitine as substrates. As expected, OVX mice presented with greater hepatic steatosis, weight gain, and fat mass gain compared with sham and OVX + Est animals. Hepatic mitochondrial coupling (basal/state 3 respiration) with pyruvate was impaired following OVX, but both VWR and estradiol treatment rescued coupling to levels greater than or equal to sham animals. Estradiol and exercise also had different effects on liver electron transport chain protein expression depending on OVX status. Markers of bile acid metabolism and excretion were also impaired by ovariectomy but rescued with estradiol add-back. Together our data suggest that estrogen depletion impairs hepatic mitochondrial function and liver health, and that estradiol replacement and modest exercise can aid in rescuing this phenotype.NEW & NOTEWORTHY OVX induces hepatic steatosis in sedentary mice which can be prevented by modest physical activity (VWR) and/or estradiol treatment. Estrogen impacts hepatic mitochondrial coupling in a substrate-specific manner. OVX mice have impaired fecal bile acid excretion, which was rescued with estradiol treatment.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
| | - Colin S McCoin
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| | - Alex T Von Schulze
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Claire J Houchen
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Michael A Choi
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Kansas Medical Center, Kansas City, Kansas
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| |
Collapse
|
46
|
Biteli P, Barbalho SM, Detregiachi CRP, Dos Santos Haber JF, Chagas EFB. Dyslipidemia influences the effect of physical exercise on inflammatory markers on obese women in post-menopause: A randomized clinical trial. Exp Gerontol 2021; 150:111355. [PMID: 33865923 DOI: 10.1016/j.exger.2021.111355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023]
Abstract
The hormonal modifications observed in post-menopausal are related to increased adiposity and alteration in the lipid profile besides physical and psychological changes. Physical exercises may attenuate these conditions and have been associated with low-grade inflammatory status, reducing the risk of cardiovascular diseases. This study aimed to evaluate the influence of dyslipidemia on the effect of physical exercise on inflammatory markers IL6, IL10, and TNF-α in obese post-menopausal women. A randomized clinical trial was carried out in seventy women divided into four groups: exercise without dyslipidemia (EG/n = 11); exercise with dyslipidemia (EGD = 24); control with dyslipidemia (CGD/n = 22); and control without dyslipidemia (CG/n = 13). The serum values of IL-6, IL-10, and TNF-α were measured before and after the intervention period, and the exercise program lasted 20 weeks, in three weekly sessions of 75 min each, with aerobic and strength exercises. The comparison of means was performed using the ANOVA test, repeated measures to analyze the interaction between the group and intervention time. There were a significant reduction in IL-6 values and an increase in IL-10/IL-6 and IL-10/TNF-α ratios only in the EG group. For serum TNF-α values, the EG and EGD groups showed significant reductions. The groups that practiced exercises did not present significant variation in the levels of IL-10. However, the CGD and GC groups showed a significant reduction in IL-10 after the intervention period.
Collapse
Affiliation(s)
- Piero Biteli
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, UNIMAR, Marília, SP, Brazil
| | - Sandra Maria Barbalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, UNIMAR, Marília, SP, Brazil; School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil; School of Food and Technology of Marilia (FATEC), Marilia, SP, Brazil.
| | | | | | | |
Collapse
|
47
|
Exercise-A Panacea of Metabolic Dysregulation in Cancer: Physiological and Molecular Insights. Int J Mol Sci 2021; 22:ijms22073469. [PMID: 33801684 PMCID: PMC8037630 DOI: 10.3390/ijms22073469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/21/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a comorbidity of many types of cancers. Disruption of glucose metabolism is of concern, as it is associated with higher cancer recurrence rates and reduced survival. Current evidence suggests many health benefits from exercise during and after cancer treatment, yet only a limited number of studies have addressed the effect of exercise on cancer-associated disruption of metabolism. In this review, we draw on studies in cells, rodents, and humans to describe the metabolic dysfunctions observed in cancer and the tissues involved. We discuss how the known effects of acute exercise and exercise training observed in healthy subjects could have a positive outcome on mechanisms in people with cancer, namely: insulin resistance, hyperlipidemia, mitochondrial dysfunction, inflammation, and cachexia. Finally, we compile the current limited knowledge of how exercise corrects metabolic control in cancer and identify unanswered questions for future research.
Collapse
|
48
|
Hydes T, Alam U, Cuthbertson DJ. The Impact of Macronutrient Intake on Non-alcoholic Fatty Liver Disease (NAFLD): Too Much Fat, Too Much Carbohydrate, or Just Too Many Calories? Front Nutr 2021; 8:640557. [PMID: 33665203 PMCID: PMC7921724 DOI: 10.3389/fnut.2021.640557] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing epidemic, in parallel with the obesity crisis, rapidly becoming one of the commonest causes of chronic liver disease worldwide. Diet and physical activity are important determinants of liver fat accumulation related to insulin resistance, dysfunctional adipose tissue, and secondary impaired lipid storage and/or increased lipolysis. While it is evident that a hypercaloric diet (an overconsumption of calories) promotes liver fat accumulation, it is also clear that the macronutrient composition can modulate this risk. A number of other baseline factors modify the overfeeding response, which may be genetic or environmental. Although it is difficult to disentangle the effects of excess calories vs. specifically the individual effects of excessive carbohydrates and/or fats, isocaloric, and hypercaloric dietary intervention studies have been implemented to provide insight into the effects of different macronutrients, sub-types and their relative balance, on the regulation of liver fat. What has emerged is that different types of fat and carbohydrates differentially influence liver fat accumulation, even when diets are isocaloric. Furthermore, distinct molecular and metabolic pathways mediate the effects of carbohydrates and fat intake on hepatic steatosis. Fat accumulation appears to act through impairments in lipid storage and/or increased lipolysis, whereas carbohydrate consumption has been shown to promote liver fat accumulation through de novo lipogenesis. Effects differ dependent upon carbohydrate and fat type. Saturated fat and fructose induce the greatest increase in intrahepatic triglycerides (IHTG), insulin resistance, and harmful ceramides compared with unsaturated fats, which have been found to be protective. Decreased intake of saturated fats and avoidance of added sugars are therefore the two most important dietary interventions that can lead to a reduction in IHTG and potentially the associated risk of developing type 2 diabetes. A healthy and balanced diet and regular physical activity must remain the cornerstones of effective lifestyle intervention to prevent the development and progression of NAFLD. Considering the sub-type of each macronutrient, in addition to the quantity, are critical determinants of liver health.
Collapse
Affiliation(s)
- Theresa Hydes
- Department of Metabolic and Cardiovascular Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Uazman Alam
- Department of Metabolic and Cardiovascular Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Daniel J Cuthbertson
- Department of Metabolic and Cardiovascular Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
49
|
Friesen CS, Hosey-Cojocari C, Chan SS, Csanaky IL, Wagner JB, Sweeney BR, Friesen A, Fraser JD, Shakhnovich V. Efficacy of Weight Reduction on Pediatric Nonalcoholic Fatty Liver Disease: Opportunities to Improve Treatment Outcomes Through Pharmacotherapy. Front Endocrinol (Lausanne) 2021; 12:663351. [PMID: 33927697 PMCID: PMC8076784 DOI: 10.3389/fendo.2021.663351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is the single greatest risk factor for nonalcoholic fatty liver disease (NAFLD). Without intervention, most pediatric patients with NAFLD continue to gain excessive weight, making early, effective weight loss intervention key for disease treatment and prevention of NAFLD progression. Unfortunately, outside of a closely monitored research setting, which is not representative of the real world, lifestyle modification success for weight loss in children is low. Bariatric surgery, though effective, is invasive and can worsen NAFLD postoperatively. Thus, there is an evolving and underutilized role for pharmacotherapy in children, both for weight reduction and NAFLD management. In this perspective article, we provide an overview of the efficacy of weight reduction on pediatric NAFLD treatment, discuss the pros and cons of currently approved pharmacotherapy options, as well as drugs commonly used off-label for weight reduction in children and adolescents. We also highlight gaps in, and opportunities for, streamlining obesity trials to include NAFLD assessment as a valuable, secondary, therapeutic outcome measure, which may aid drug repurposing. Finally, we describe the already available, and emerging, minimally-invasive biomarkers of NAFLD that could offer a safe and convenient alternative to liver biopsy in pediatric obesity and NAFLD trials.
Collapse
Affiliation(s)
- Chance S. Friesen
- University of Kansas School of Medicine, Kansas City, KS, United States
| | | | - Sherwin S. Chan
- Children’s Mercy Kansas City, Kansas City, MO, United States
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Iván L. Csanaky
- Children’s Mercy Kansas City, Kansas City, MO, United States
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- University of Kansas Medical Center, Kansas City, KS, United States
| | - Jonathan B. Wagner
- Children’s Mercy Kansas City, Kansas City, MO, United States
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Brooke R. Sweeney
- Children’s Mercy Kansas City, Kansas City, MO, United States
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Center for Children’s Healthy Lifestyles & Nutrition, Kansas City, MO, United States
| | - Alec Friesen
- University of Kansas School of Medicine, Kansas City, KS, United States
| | - Jason D. Fraser
- Children’s Mercy Kansas City, Kansas City, MO, United States
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Valentina Shakhnovich
- Children’s Mercy Kansas City, Kansas City, MO, United States
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- University of Kansas Medical Center, Kansas City, KS, United States
- Center for Children’s Healthy Lifestyles & Nutrition, Kansas City, MO, United States
- *Correspondence: Valentina Shakhnovich,
| |
Collapse
|
50
|
Plaza-Díaz J, Solís-Urra P, Rodríguez-Rodríguez F, Olivares-Arancibia J, Navarro-Oliveros M, Abadía-Molina F, Álvarez-Mercado AI. The Gut Barrier, Intestinal Microbiota, and Liver Disease: Molecular Mechanisms and Strategies to Manage. Int J Mol Sci 2020; 21:E8351. [PMID: 33171747 PMCID: PMC7664383 DOI: 10.3390/ijms21218351] [Citation(s) in RCA: 64] [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: 09/28/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Liver disease encompasses pathologies as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, alcohol liver disease, hepatocellular carcinoma, viral hepatitis, and autoimmune hepatitis. Nowadays, underlying mechanisms associating gut permeability and liver disease development are not well understood, although evidence points to the involvement of intestinal microbiota and their metabolites. Animal studies have shown alterations in Toll-like receptor signaling related to the leaky gut syndrome by the action of bacterial lipopolysaccharide. In humans, modifications of the intestinal microbiota in intestinal permeability have also been related to liver disease. Some of these changes were observed in bacterial species belonging Roseburia, Streptococcus, and Rothia. Currently, numerous strategies to treat liver disease are being assessed. This review summarizes and discusses studies addressed to determine mechanisms associated with the microbiota able to alter the intestinal barrier complementing the progress and advancement of liver disease, as well as the main strategies under development to manage these pathologies. We highlight those approaches that have shown improvement in intestinal microbiota and barrier function, namely lifestyle changes (diet and physical activity) and probiotics intervention. Nevertheless, knowledge about how such modifications are beneficial is still limited and specific mechanisms involved are not clear. Thus, further in-vitro, animal, and human studies are needed.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada;
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
| | - Patricio Solís-Urra
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2531015, Chile;
| | - Fernando Rodríguez-Rodríguez
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile; (F.R.-R.); (J.O.-A.)
| | - Jorge Olivares-Arancibia
- IRyS Research Group, School of Physical Education, Pontificia Universidad Católica de Valparaíso, Valparaíso 2374631, Chile; (F.R.-R.); (J.O.-A.)
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad de las Américas, Santiago 8370035, Chile
| | - Miguel Navarro-Oliveros
- BioCritic. Group for Biomedical Research in Critical Care Medicine, 47005 Valladolid, Spain;
| | - Francisco Abadía-Molina
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain;
- Department of Cell Biology, School of Sciences, University of Granada, 18071 Granada, Spain
| | - Ana I. Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain;
| |
Collapse
|