1
|
Jamerlan AM, An SSA, Hulme JP. Microbial diversity and fitness in the gut-brain axis: influences on developmental risk for Alzheimer's disease. Gut Microbes 2025; 17:2486518. [PMID: 40207973 PMCID: PMC11988266 DOI: 10.1080/19490976.2025.2486518] [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: 08/28/2024] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
The gut-brain axis (GBA) denotes the dynamic and bidirectional communication system that connects the gastrointestinal tract and the central nervous system (CNS). This review explored this axis, focusing on the role of microbial diversity and fitness in maintaining gastrointestinal health and preventing neurodegeneration, particularly in Alzheimer's disease (AD). Gut dysbiosis, characterized by the imbalance in populations of beneficial and harmful bacteria, has been associated with increased systemic inflammation, neuroinflammation, and the progression of AD through pathogenic mechanisms involving amyloid deposition, tauopathy, and increased blood-brain barrier (BBB) permeability. Emerging evidence highlighted the therapeutic potential of probiotics, dietary interventions, and intermittent fasting in restoring microbial balance, reducing inflammation, and minimizing neurodegenerative risks. Probiotics and synbiotics are promising in helping improve cognitive function and metabolic health, while dietary patterns like the Mediterranean diet were linked to decreased neuroinflammation and enhanced gut-brain communication. Despite significant advancement, further research is needed to elucidate the specific microbial strains, metabolites, and mechanisms influencing brain health. Future studies employing longitudinal designs and advanced omics technologies are essential to developing targeted microbiome-based therapies for managing AD-related disorders.
Collapse
Affiliation(s)
- Angelo M. Jamerlan
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| | - Seong Soo A. An
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| | - John P. Hulme
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| |
Collapse
|
2
|
Gipson EZ, Deru LS, Graves PG, Jacobsen CG, Peterson NE, Bailey BW. The effects of initiating a 24-hour fast with a low versus a high carbohydrate shake on glycemic control in older adults: a randomized crossover study. Nutr Metab (Lond) 2025; 22:32. [PMID: 40234969 PMCID: PMC11998415 DOI: 10.1186/s12986-025-00920-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/19/2025] [Indexed: 04/17/2025] Open
Abstract
PURPOSE This study measured the impact of macronutrient composition of a pre-fast meal on time-to-ketosis and other metabolic indicators of glycemic control during a 24-hr fast within a population of older, sedentary, overweight adults. METHODS Twenty-four adults who were over the age of 50, sedentary (< 150 min of weekly exercise), and overweight (BMI ≥ 27) participated in a randomized crossover study. Each of these inclusion criteria have been shown to increase the risk for the development of chronic diseases. Participants began each 24-hr fast with either a high carbohydrate/low fat/moderate protein (HC/LF) or an isocaloric low carbohydrate/high fat/moderate protein (LC/HF) shake. Metabolic indicators included subcutaneous glucose readings every 15 min throughout the study, capillary beta-hydroxybutyrate (BHB), and plasma concentrations of insulin, glucagon, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP). Measurements of these hormones took place at 0, 1, 24, and 48 h, and BHB measurements took place at 0, 1, 4, 8, 12 and 24 h. RESULTS Glucose levels were higher in the HC/LF group 15 min to 2.25 h after fast initiation (p < 0.05 for all). There was a significant condition by time interaction for BHB (F = 3.84, p < 0.01). Nutritional ketosis (BHB ≥ 0.5 mmol/L) was reached on average by 12 h in the LC/HF condition but was not reached at any point during the fast on average in the HC/LF condition. An hour after consuming the LC/HF shake insulin was 41.9% lower (t = 6.13, p < 0.01), glucagon 23.6% higher (t = -4.72, p < 0.01), GLP-1 26.8% higher (t = -5.16, p < 0.01), and GIP 34.4% higher (t = -3.41, p < 0.01) compared to the HC/LF shake. CONCLUSION A low carbohydrate pre-fast meal can reduce time-to-ketosis in older, sedentary, overweight adults. Those looking to improve glycemic control through fasting or time-restricted eating interventions may consider the macronutrient composition of their pre-fast meal to improve its efficacy.
Collapse
Affiliation(s)
- Elizabeth Z Gipson
- Department of Exercise Science, Brigham Young University, Provo, UT, 84602, USA.
| | - Landon S Deru
- Department of Exercise Science, Brigham Young University, Provo, UT, 84602, USA
- Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Parker G Graves
- Department of Exercise Science, Brigham Young University, Provo, UT, 84602, USA
| | - Cameron G Jacobsen
- Department of Exercise Science, Brigham Young University, Provo, UT, 84602, USA
| | - Neil E Peterson
- College of Nursing, Brigham Young University, Provo, UT, 84602, USA
| | - Bruce W Bailey
- Department of Exercise Science, Brigham Young University, Provo, UT, 84602, USA
| |
Collapse
|
3
|
Beli E, Yan Y, Moldovan L, Lydic TA, Krishman P, Tersey SA, Duan Y, Salazar TE, Dominguez JM, Nguyen DV, Cox A, Li Calzi S, Beam C, Mirmira RG, Evans-Molina C, Busik JV, Grant MB. Reshaping lipid metabolism with long-term alternate day feeding in type 2 diabetes mice. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:3. [PMID: 39911696 PMCID: PMC11790504 DOI: 10.1038/s44324-024-00039-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/07/2024] [Indexed: 02/07/2025]
Abstract
Strategies to improve metabolic health include calorie restriction, time restricted eating and fasting several days per week or month. These approaches have demonstrated benefits for individuals experiencing obesity, metabolic syndrome, and prediabetes. However, their impact on established diabetes remains incompletely studied. The chronicity of type 2 diabetes (T2D) requires that interventions must be undertaken for extended periods of time, typically the entire lifetime of the individual. In this study, we examined the impact of intermittent fasting (IF), with an every-other-day protocol for a duration of 6 months in a murine model of T2D, the db/db (D) mouse on metabolism and liver steatosis. We compared D-IF mice with diabetic ad-libitum (AL; D-AL), control-IF (C-IF) and control-AL (C-AL) cohorts. We demonstrated using lipidomic, microbiome, metabolomic and liver transcriptomic studies that chronic IF improved carbohydrate utilization and glucose homeostasis without weight loss and reduced white adipose tissue inflammation and significantly impacted lipid metabolism in the liver. Microbiome studies and predicted functional analysis of gut microbiota showed that IF increased beneficial bacteria involved in sphingolipid (SL) metabolism. The metabolomic studies showed that oxidation of lipid species and ceramide levels were reduced in D-IF compared to D-AL. The liver lipidomic analysis and liver microarray confirmed a reduction in overall lipid content in D-IF mice compared to D-AL mice, especially in the feeding state as well as an overall reduction in oxidized lipids and ceramides. These studies support that long-term IF can improve glucose homeostasis and dramatically altered lipid metabolism in the absence of weight loss.
Collapse
Affiliation(s)
- Eleni Beli
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
- Center for Diabetes and Metabolic Diseases, and the Herman B Wells
Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
- Present Address: Wellcome-Wolfson Institute of Experimental Medicine, Queen’s University Belfast, Belfast, UK
| | - Yuanqing Yan
- Department of Surgery, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leni Moldovan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
- Present Address: Department of Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| | - Todd A. Lydic
- Department of Physiology, Michigan State University, East Lansing, MI USA
| | - Preethi Krishman
- Center for Diabetes and Metabolic Diseases, and the Herman B Wells
Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
| | - Sarah A. Tersey
- Center for Diabetes and Metabolic Diseases, and the Herman B Wells
Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
- Present Address: Department of Medicine, The University of Chicago, Chicago, IL USA
| | - Yaqian Duan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Tatiana E. Salazar
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
| | - James M. Dominguez
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Dung V. Nguyen
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Abigail Cox
- Department of Comparative Pathobiology, College of Veterinary
Medicine, Purdue University, Lafayette, IN USA
| | - Sergio Li Calzi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
- Present Address: Department of Ophthalmology and Visual Science, University of Alabama Birmingham, Birmingham, AL USA
| | - Craig Beam
- Department of Biomedical Sciences, Homer Stryker MD School of
Medicine, Western Michigan University, Kalamazoo, MI USA
| | - Raghavendra G. Mirmira
- Center for Diabetes and Metabolic Diseases, and the Herman B Wells
Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
- Present Address: Department of Medicine, The University of Chicago, Chicago, IL USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, and the Herman B Wells
Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
| | - Julia V. Busik
- Department of Physiology, Michigan State University, East Lansing, MI USA
- Present Address: Department of Biochemistry and Physiology, The university of Oklahoma Health Sciences, Oklahoma City, OK USA
| | - Maria B. Grant
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN USA
- Present Address: Department of Ophthalmology and Visual Science, University of Alabama Birmingham, Birmingham, AL USA
| |
Collapse
|
4
|
Xu G, Qiu F, Zhang W, Li S, Chen J, Wang G, Wang Y, Pan J, Pan X. Fibroblast growth factor 21 alleviates acetaminophen induced acute liver injury by activating Sirt1 mediated autophagy. Cell Signal 2024; 123:111379. [PMID: 39233207 DOI: 10.1016/j.cellsig.2024.111379] [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: 05/04/2024] [Revised: 07/27/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND AND AIMS Acetaminophen (APAP) is the main cause of acute liver injury (ALI) in the Western. Our previous study has shown that fenofibrate activated hepatic expression of fibroblast growth factor 21 (FGF21) can protect the liver form APAP injuries by promoting autophagy. However, the underlying mechanism involved in FGF21-mediated autophagy remains unsolved. METHODS The ALI mice model was established by intraperitoneal injection of APAP. To investigate the influence of FGF21 on autophagy and Sirt1 expression in APAP-induced ALI, FGF21 knockout (FGF21KO) mice and exogenously supplemented mouse recombinant FGF21 protein were used. In addition, primary isolated hepatocytes and the Sirt1 inhibitor EX527 were used to observe whether FGF21 activated autophagy in APAP injury is regulated by Sirt1 at the cellular level. RESULTS FGF21, Sirt1, and autophagy levels increased in mice with acute liver injury (ALI) and in primary cultured hepatocytes. Deletion of the FGF21 gene exacerbated APAP-induced liver necrosis and oxidative stress, and decreased mitochondrial potential. It also reduced the mRNA and protein levels of autophagy-related proteins such as Sirt1, LC3-II, and p62, as well as the number of autophagosomes. Replenishment of FGF21 reversed these processes. In addition, EX527 partially counteracted the protective effect of FGF21 by worsening oxidative damage, mitochondrial damage, and reducing autophagy in primary liver cells treated with APAP. CONCLUSION FGF21 increases autophagy by upregulating Sirt1 to alleviate APAP-induced injuries.
Collapse
Affiliation(s)
- Guangsen Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Feng Qiu
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Wenshu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China; Department of Pharmacy, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang 311100, China
| | - Supeng Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Jiale Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Guiyun Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Ye Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xuebo Pan
- School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
5
|
Zhang A, Wang J, Zhao Y, He Y, Sun N. Intermittent fasting, fatty acid metabolism reprogramming, and neuroimmuno microenvironment: mechanisms and application prospects. Front Nutr 2024; 11:1485632. [PMID: 39512520 PMCID: PMC11541237 DOI: 10.3389/fnut.2024.1485632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Intermittent fasting (IF) has demonstrated extensive health benefits through the regulation of fatty acid metabolism and modulation of the neuroimmune microenvironment, primarily via the activation of key signaling pathways such as AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1). IF not only facilitates fatty acid oxidation and improves metabolic health, but also enhances mitochondrial function, mitigates oxidative stress, promotes autophagy, and inhibits apoptosis and ferroptosis. These mechanisms contribute to its substantial preventive and therapeutic potential in various conditions, including neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, autoimmune diseases, and neurotraumatic conditions. While supportive evidence has been obtained from animal models and preliminary clinical studies, further large-scale, long-term randomized controlled trials are imperative to establish its safety and evaluate its clinical efficacy comprehensively.
Collapse
Affiliation(s)
- Anren Zhang
- Department of Rehabilitation, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junyu Wang
- Department of Rehabilitation, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinuo Zhao
- Department of Rehabilitation, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu He
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Nianyi Sun
- Department of Rehabilitation, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Zheng D, Hong X, He X, Lin J, Fan S, Wu J, Liang Z, Chen S, Yan L, Ren M, Wang W. Intermittent Fasting-Improved Glucose Homeostasis Is Not Entirely Dependent on Caloric Restriction in db/db Male Mice. Diabetes 2024; 73:864-878. [PMID: 38502858 PMCID: PMC11109801 DOI: 10.2337/db23-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
Intermittent fasting (IF), which involves prolonged fasting intervals accompanied by caloric restriction (CR), is an effective dietary treatment for obesity and diabetes. Although IF offers many benefits, it is difficult to determine whether these benefits are the consequences of CR. Every-other-day feeding (EODF) is a commonly used IF research model. This study was designed to identify factors, in addition to CR, responsible for the effects of EODF and the possible underlying mechanisms. Diabetic db/db mice were divided into three groups: ad libitum (AL), meal feeding (MF), and EODF. The MF model was used to attain a level of CR comparable to that of EODF, with food distribution evenly divided between 10:00 a.m. and 6:00 p.m., thereby minimizing the fasting interval. EODF yielded greater improvements in glucose homeostasis than MF in db/db mice by reducing fasting glucose levels and enhancing glucose tolerance. However, these effects on glucose metabolism were less pronounced in lean mice. Furthermore, ubiquitination of the liver-specific glucocorticoid (GC) receptor (GR) facilitated its degradation and downregulation of Kruppel-like factor 9 (KLF9), which ultimately suppressed liver gluconeogenesis in diabetic EODF mice. Although GR and KLF9 might mediate the metabolic benefits of EODF, the potential benefits of EODF might be limited by elevated serum GC levels in diabetic EODF mice. Overall, this study suggests that the metabolic benefits of EODF in improving glucose homeostasis are independent of CR, possibly because of the downstream effects of liver-specific GR degradation. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Dinghao Zheng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Xiaosi Hong
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Xiaodan He
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Jianghong Lin
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Shujin Fan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Jinli Wu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Zhuoxian Liang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Li Yan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Meng Ren
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
| | - Wei Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou, China
- Department of Endocrinology, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Deng Y, Yang X, Ye X, Yuan Y, Zhang Y, Teng F, You D, Zhou X, Liu W, Li K, Luo S, Yang Z, Chen R, Shi G, Li J, Zhang H. Alternate day fasting aggravates atherosclerosis through the suppression of hepatic ATF3 in Apoe-/- mice. LIFE METABOLISM 2024; 3:loae009. [PMID: 39872376 PMCID: PMC11749235 DOI: 10.1093/lifemeta/loae009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 01/30/2025]
Abstract
Atherosclerosis is the major contributor to cardiovascular mortality worldwide. Alternate day fasting (ADF) has gained growing attention due to its metabolic benefits. However, the effects of ADF on atherosclerotic plaque formation remain inconsistent and controversial in atherosclerotic animal models. The present study was designed to investigate the effects of ADF on atherosclerosis in apolipoprotein E-deficient (Apoe -/- ) mice. Eleven-week-old male Apoe -/- mice fed with Western diet (WD) were randomly grouped into ad libitum (AL) group and ADF group, and ADF aggravated both the early and advanced atherosclerotic lesion formation, which might be due to the disturbed cholesterol profiles caused by ADF intervention. ADF significantly altered cholesterol metabolism pathways and down-regulated integrated stress response (ISR) in the liver. The hepatic expression of activating transcription factor 3 (ATF3) was suppressed in mice treated with ADF and hepatocyte-specific overexpression of Aft3 attenuated the effects of ADF on atherosclerotic plaque formation in Apoe -/- mice. Moreover, the expression of ATF3 could be regulated by Krüppel-like factor 6 (KLF6) and both the expressions of ATF3 and KLF6 were regulated by hepatic cellular ISR pathway. In conclusion, ADF aggravates atherosclerosis progression in Apoe -/- mice fed on WD. ADF inhibits the hepatic ISR signaling pathway and decreases the expression of KLF6, subsequently inhibiting ATF3 expression. The suppressed ATF3 expression in the liver mediates the deteriorated effects of ADF on atherosclerosis in Apoe -/- mice. The findings suggest the potentially harmful effects when ADF intervention is applied to the population at high risk of atherosclerosis.
Collapse
Affiliation(s)
- Yajuan Deng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaoyu Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xueru Ye
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Youwen Yuan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yanan Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fei Teng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Danming You
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xuan Zhou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wenhui Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kangli Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shenjian Luo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhi Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ruxin Chen
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Guojun Shi
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Jin Li
- Department of Endocrinology, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
8
|
Shen W, Yang M, Chen H, He C, Li H, Yang X, Zhuo J, Lin Z, Hu Z, Lu D, Xu X. FGF21-mediated autophagy: Remodeling the homeostasis in response to stress in liver diseases. Genes Dis 2024; 11:101027. [PMID: 38292187 PMCID: PMC10825283 DOI: 10.1016/j.gendis.2023.05.019] [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: 02/24/2023] [Revised: 04/23/2023] [Accepted: 05/09/2023] [Indexed: 02/01/2024] Open
Abstract
Liver diseases are worldwide problems closely associated with various stresses, such as endoplasmic reticulum stress. The exact interplay between stress and liver diseases remains unclear. Autophagy plays an essential role in maintaining homeostasis, and recent studies indicate tight crosstalk between stress and autophagy in liver diseases. Once the balance between damage and autophagy is broken, autophagy can no longer resist injury or maintain homeostasis. In recent years, FGF21 (fibroblast growth factor 21)-induced autophagy has attracted much attention. FGF21 is regarded as a stress hormone and can be up-regulated by an abundance of signaling pathways in response to stress. Also, increased FGF21 activates autophagy by a complicated signaling network in which mTOR plays a pivotal role. This review summarizes the mechanism of FGF21-mediated autophagy and its derived application in the defense of stress in liver diseases and offers a glimpse into its promising prospect in future clinical practice.
Collapse
Affiliation(s)
- Wei Shen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Modan Yang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Hao Chen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Chiyu He
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Huigang Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xinyu Yang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Jianyong Zhuo
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zuyuan Lin
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zhihang Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Di Lu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xiao Xu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- The Institute for Organ Repair and Regenerative Medicine of Hangzhou, Hangzhou, Zhejiang 310006, China
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, Zhejiang 310003, China
- National Center for Healthcare Quality Management in Liver Transplant, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
9
|
Wells RG, Neilson LE, McHill AW, Hiller AL. Dietary fasting and time-restricted eating in Huntington's disease: therapeutic potential and underlying mechanisms. Transl Neurodegener 2024; 13:17. [PMID: 38561866 PMCID: PMC10986006 DOI: 10.1186/s40035-024-00406-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder caused by aggregation of the mutant huntingtin (mHTT) protein, resulting from a CAG repeat expansion in the huntingtin gene HTT. HD is characterized by a variety of debilitating symptoms including involuntary movements, cognitive impairment, and psychiatric disturbances. Despite considerable efforts, effective disease-modifying treatments for HD remain elusive, necessitating exploration of novel therapeutic approaches, including lifestyle modifications that could delay symptom onset and disease progression. Recent studies suggest that time-restricted eating (TRE), a form of intermittent fasting involving daily caloric intake within a limited time window, may hold promise in the treatment of neurodegenerative diseases, including HD. TRE has been shown to improve mitochondrial function, upregulate autophagy, reduce oxidative stress, regulate the sleep-wake cycle, and enhance cognitive function. In this review, we explore the potential therapeutic role of TRE in HD, focusing on its underlying physiological mechanisms. We discuss how TRE might enhance the clearance of mHTT, recover striatal brain-derived neurotrophic factor levels, improve mitochondrial function and stress-response pathways, and synchronize circadian rhythm activity. Understanding these mechanisms is critical for the development of targeted lifestyle interventions to mitigate HD pathology and improve patient outcomes. While the potential benefits of TRE in HD animal models are encouraging, future comprehensive clinical trials will be necessary to evaluate its safety, feasibility, and efficacy in persons with HD.
Collapse
Affiliation(s)
- Russell G Wells
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Lee E Neilson
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Neurology and PADRECC VA Portland Health Care System, Portland, OR, 97239, USA
| | - Andrew W McHill
- Sleep, Chronobiology and Health Laboratory, School of Nursing, Oregon Health & Science University, Portland, OR, 97239, USA
- Oregon Institute of Occupational Health Sciences, Oregon Health & Sciences University, Portland, OR, 97239, USA
| | - Amie L Hiller
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Neurology and PADRECC VA Portland Health Care System, Portland, OR, 97239, USA
| |
Collapse
|
10
|
Gao Y, Zhai W, Sun L, Du X, Wang X, Mulholland MW, Yin Y, Zhang W. Hepatic LGR4 aggravates cholestasis-induced liver injury in mice. Am J Physiol Gastrointest Liver Physiol 2024; 326:G460-G472. [PMID: 38440827 PMCID: PMC11213478 DOI: 10.1152/ajpgi.00127.2023] [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: 06/27/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
Current therapy for hepatic injury induced by the accumulation of bile acids is limited. Leucine-rich repeat G protein-coupled receptor 4 (LGR4), also known as GPR48, is critical for cytoprotection and cell proliferation. Here, we reported a novel function for the LGR4 in cholestatic liver injury. In the bile duct ligation (BDL)-induced liver injury model, hepatic LGR4 expression was significantly downregulated. Deficiency of LGR4 in hepatocytes (Lgr4LKO) notably decreased BDL-induced liver injury measured by hepatic necrosis, fibrosis, and circulating liver enzymes and total bilirubin. Levels of total bile acids in plasma and liver were markedly reduced in these mice. However, deficiency of LGR4 in macrophages (Lyz2-Lgr4MKO) demonstrated no significant effect on liver injury induced by BDL. Deficiency of LGR4 in hepatocytes significantly attenuated S1PR2 and the phosphorylation of protein kinase B (AKT) induced by BDL. Recombinant Rspo1 and Rspo3 potentiated the taurocholic acid (TCA)-induced upregulation in S1PR2 and phosphorylation of AKT in hepatocytes. Inhibition of S1PR2-AKT signaling by specific AKT or S1PR2 inhibitors blocked the increase of bile acid secretion induced by Rspo1/3 in hepatocytes. Our studies indicate that the R-spondins (Rspos)-LGR4 signaling in hepatocytes aggravates the cholestatic liver injury by potentiating the production of bile acids in a S1PR2-AKT-dependent manner.NEW & NOTEWORTHY Deficiency of LGR4 in hepatocytes alleviates BDL-induced liver injury. LGR4 in macrophages demonstrates no effect on BDL-induced liver injury. Rspos-LGR4 increases bile acid synthesis and transport via potentiating S1PR2-AKT signaling in hepatocytes.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
| | - Wenbo Zhai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
| | - Xueqian Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
| | - Xianfeng Wang
- Department of Pharmacology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
| | - Michael W Mulholland
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, United States
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, People's Republic of China
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, United States
| |
Collapse
|
11
|
Mirrazavi ZS, Behrouz V. Various types of fasting diet and possible benefits in nonalcoholic fatty liver: Mechanism of actions and literature update. Clin Nutr 2024; 43:519-533. [PMID: 38219703 DOI: 10.1016/j.clnu.2024.01.005] [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: 10/22/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the major causes of chronic liver injury, affecting around one-fourth of the general population across the world. Several important pathophysiological mechanisms underlying NAFLD include oxidative stress, inflammation, liver fibrosis, and apoptosis. Currently, therapeutic approaches are not ideal for managing NAFLD, thus new approaches and treatments are still needed. Over the last two decades, various fasting protocols have been explored to reduce body weight and improve metabolic disorders. In this review, we provide updated literature that supports fasting regimens for subjects with NAFLD and describes underlying mechanisms of action. We suggest that fasting regimens may modulate NAFLD via several mechanisms, including changes in gut microbiota, hepatic arginase, hepatic autophagy, inflammatory responses, liver functional enzymes and hepatic steatosis, fibroblast growth factors signaling, white adipose tissue browning, adipokines, circadian rhythms, lipid profiles, and body composition.
Collapse
Affiliation(s)
| | - Vahideh Behrouz
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
12
|
Yu H, Zhong D, Li S, Mo H, Zhang Z, Gao J, Ren X, Yu J, Geng S, Wang Y, Li Y, Wang L. FGF21 Improves Glycolipid Metabolism in Rainbow Trout ( Oncorhynchus mykiss) Fed a High-Carbohydrate Diet by Inhibiting Inflammatory Responses and Activating Autophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20118-20130. [PMID: 38061326 DOI: 10.1021/acs.jafc.3c06768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
In this study, the coding region of rainbow trout fgf21 was cloned and sequenced to synthesize a recombinant protein (rFGF21) and investigate its potential role in improving glycolipid metabolism. Acute injection of rFGF21 into rainbow trout effectively reduced serum glucose levels. To investigate the effect of rFGF21 on high-carbohydrate diet (HCD)-induced metabolic disorders in rainbow trout, a 31-day feeding experiment was conducted. At the end of the third week, fish were injected with either PBS or rFGF21. The results showed that the final body weight (FBW) significantly increased in rainbow trout on an HCD (P < 0.05), but there were potential risks including disturbances in glycolipid metabolism and increased inflammatory responses. However, these effects were altered by rFGF21 treatment. In addition, rFGF21 promotes glucose uptake by increasing the phosphorylation levels of AKT (protein kinase B) and GSK3β (glycogen synthase kinase 3β), increasing hepatic glycogen, thereby lowering serum glucose. Notably, the rFGF21 did not exacerbate the inflammatory response but downregulated the expression of inflammatory factors. Interestingly, the activation of autophagy and the AMPK pathway may contribute to the positive effect of rFGF21, where rFGF21 injection significantly increased the levels of LC3I/II protein and phosphorylate AMPKα (P < 0.05).
Collapse
Affiliation(s)
- Huixia Yu
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Debin Zhong
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Shuai Li
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Haolin Mo
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Zhihao Zhang
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Jiuwei Gao
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Xin Ren
- Meixian Aquaculture Farm of Shitouhe Reservoir Administration, Xianyang 712000, Shaanxi, China
| | - Jiajia Yu
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Shuo Geng
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Yingwei Wang
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Yang Li
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| | - Lixin Wang
- College of Animal Science and Technology, Northwest A & F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
13
|
Yin J, Cheng L, Hong Y, Li Z, Li C, Ban X, Zhu L, Gu Z. A Comprehensive Review of the Effects of Glycemic Carbohydrates on the Neurocognitive Functions Based on Gut Microenvironment Regulation and Glycemic Fluctuation Control. Nutrients 2023; 15:5080. [PMID: 38140339 PMCID: PMC10745758 DOI: 10.3390/nu15245080] [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: 11/15/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Improper glycemic carbohydrates (GCs) consumption can be a potential risk factor for metabolic diseases such as obesity and diabetes, which may lead to cognitive impairment. Although several potential mechanisms have been studied, the biological relationship between carbohydrate consumption and neurocognitive impairment is still uncertain. In this review, the main effects and mechanisms of GCs' digestive characteristics on cognitive functions are comprehensively elucidated. Additionally, healthier carbohydrate selection, a reliable research model, and future directions are discussed. Individuals in their early and late lives and patients with metabolic diseases are highly susceptible to dietary-induced cognitive impairment. It is well known that gut function is closely related to dietary patterns. Unhealthy carbohydrate diet-induced gut microenvironment disorders negatively impact cognitive functions through the gut-brain axis. Moreover, severe glycemic fluctuations, due to rapidly digestible carbohydrate consumption or metabolic diseases, can impair neurocognitive functions by disrupting glucose metabolism, dysregulating calcium homeostasis, oxidative stress, inflammatory responses, and accumulating advanced glycation end products. Unstable glycemic status can lead to more severe neurological impairment than persistent hyperglycemia. Slow-digested or resistant carbohydrates might contribute to better neurocognitive functions due to stable glycemic response and healthier gut functions than fully gelatinized starch and nutritive sugars.
Collapse
Affiliation(s)
- Jian Yin
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
| | - Li Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Zhaofeng Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Caiming Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Xiaofeng Ban
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Ling Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (J.Y.); (Y.H.); (Z.L.); (C.L.); (X.B.); (L.Z.)
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
14
|
Li Z, Huang L, Luo Y, Yu B, Tian G. Effects and possible mechanisms of intermittent fasting on health and disease: a narrative review. Nutr Rev 2023; 81:1626-1635. [PMID: 36940184 DOI: 10.1093/nutrit/nuad026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
The imbalance between energy intake and expenditure in an environment of continuous food availability can lead to metabolic disturbances in the body and increase the risk of obesity and a range of chronic noncommunicable diseases. Intermittent fasting (IF) is one of the most popular nonpharmacological interventions to combat obesity and chronic noncommunicable diseases. The 3 most widely studied IF regimens are alternate-day fasting, time-restricted feeding, and the 5:2 diet. In rodents, IF helps optimize energy metabolism, prevent obesity, promote brain health, improve immune and reproductive function, and delay aging. In humans, IF's benefits are relevant for the aging global population and for increasing human life expectancy. However, the optimal model of IF remains unclear. In this review, the possible mechanisms of IF are summarized and its possible drawbacks are discussed on the basis of the results of existing research, which provide a new idea for nonpharmaceutical dietary intervention of chronic noncommunicable diseases.
Collapse
Affiliation(s)
- Zimei Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Liansu Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Yuheng Luo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Bing Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Gang Tian
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| |
Collapse
|
15
|
Li D, Dun Y, Qi D, Ripley-Gonzalez JW, Dong J, Zhou N, Qiu L, Zhang J, Zeng T, You B, Liu S. Intermittent fasting activates macrophage migration inhibitory factor and alleviates high-fat diet-induced nonalcoholic fatty liver disease. Sci Rep 2023; 13:13068. [PMID: 37567977 PMCID: PMC10421944 DOI: 10.1038/s41598-023-40373-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
Switching to normal diet (ND) is the regular therapy for high-fat diet (HFD)-induced nonalcoholic fatty liver disease (NAFLD). Intermittent fasting (IF) is a unique treatment which may exhibits better therapeutic efficacy. Thus, we aim to investigate the therapeutic effects of these treatments and exploring the mechanisms. In the present study, NAFLD mouse model was induced by a 10-week HFD. Thereafter, mice adopted continued HFD, ND, or IF for the next 12 weeks. Finally, the liver was then harvested to assess lipid deposition, lipid metabolism, apoptosis, and autophagy, while blood was collected to determine blood glucose and insulin. The results showed that IF and ND treatment improved lipid deposition and metabolic disorder of NAFLD mice; the increasing body weight, liver weight, and HOMA-IR index of HFD mice were also alleviated by IF and ND. Furthermore, IF and ND treatment activated the macrophage migration inhibitory factor (MIF)/AMPK pathway and regulated its downstream autophagy and apoptosis. However, the efficacy of IF was better than ND. Both IF and ND activates MIF signaling and alleviate the lipotoxicity of NAFLD while IF therapy is more effective than ND. The different MIF up-regulation might be the underlying mechanism of why IF benefits more than ND.
Collapse
Affiliation(s)
- Dezhao Li
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Yaoshan Dun
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Dake Qi
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Jeffrey W Ripley-Gonzalez
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Jie Dong
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Nanjiang Zhou
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Ling Qiu
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Jie Zhang
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Tanghao Zeng
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Baiyang You
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.
| | - Suixin Liu
- Division of Cardiac Rehabilitation, Department of Physical Medicine and Rehabilitation, Xiangya Hospital Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, Hunan, China.
| |
Collapse
|
16
|
Xiong F, Jiang K, Wu Y, Lou C, Ding C, Zhang W, Zhang X, Li C, Zheng H, Gao H. Intermittent fasting alleviates type 1 diabetes-induced cognitive dysfunction by improving the frontal cortical metabolic disorder. Biochim Biophys Acta Mol Basis Dis 2023:166725. [PMID: 37127173 DOI: 10.1016/j.bbadis.2023.166725] [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: 01/20/2023] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Intermittent fasting (IF) is an ecological strategy to control various metabolic disorder symptoms, but its protective effect on type 1 diabetes (T1D)-induced cognitive dysfunction and the underlying mechanisms remain poorly defined. Herein, we examined the efficacy of IF in altering the behaviors and brain metabolome in T1D mice and investigated the potential molecular mechanisms. We demonstrated that IF remarkably improved frontal cortical-dependent memory in T1D mice and reduced the loss of neuronal cells. Metabolomics and targeted mass spectrometry assay showed that IF reprogrammed the frontal cortical metabolome composition, including activated the aspartate and glutamate pathway and reversed glycerophospholipid and sphingolipid depositions in T1D mice. Mechanistically, IF attenuated the levels of oxidative stress proteins, such as NOX2, NOX4, 8-OHdG, 4-HNE, and inhibited the levels of pro-apoptotic factors Bax and cleaved Caspase-3, finally improved the memory ability of T1D mice. In vitro studies confirmed the protective effect of the supplemented N-acetylaspartate, a pivotal metabolite involved in IF-regulated T1D-induced cognitive dysfunction, in high glucose-stimulated SH-SY5Y cells by eliminating toxic lipids accumulation, oxidative stress and apoptosis. To conclude, the frontal cortical metabolites mediated the protective effects of IF against T1D-induced cognitive dysfunction by attenuating oxidative stress and apoptotic signaling. Thus, IF can be a potential therapeutic strategy for T1D-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Fen Xiong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Kaiyuan Jiang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yali Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Cong Lou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Chengjie Ding
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenli Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xi Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Chen Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hong Zheng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hongchang Gao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou 325035, China.
| |
Collapse
|
17
|
Liu L, Zhuo Y, Zhang H, Li J, Jiang X, Han X, Chao J, Feng B, Che L, Xu S, Lin Y, Li J, Fang Z, Sun M, Luo T, Wu D, Hua L. Time-restricted feeding ameliorates uterine epithelial estrogen receptor α transcriptional activity at the time of embryo implantation in mice fed a high-fat diet. J Nutr 2023:S0022-3166(23)37555-2. [PMID: 37062485 DOI: 10.1016/j.tjnut.2023.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND More than 30% of reproductive-age women are obese or overweight. Obesity and exposure to a high-fat diet (HFD) detrimentally affect endometrial development and embryo implantation. We previously reported that time-restricted feeding (TRF) improved ovarian follicular development, but whether and how TRF modulates embryo implantation are poorly understood. OBJECTIVE We investigated the effect of TRF on embryo implantation. METHODS In TRF group, mice had 10 hours of food free access from 9 pm to 7 am, and fed a normal diet or a HFD. Tail vein injection of Chicago blue dye was used to examine embryo implantation sites at day 5.5 (D5.5) of pregnancy. Serum collected at D0.5 and D4.5 of pregnancy was used to examine the level of estradiol (E2) and progesterone. Uterine estrogen receptor (ER) and progesterone receptor levels and their targeted aquaporins (AQPs) were measured. LC-MS was used to analyze bile acid (BA) composition, and primary hepatocytes were used to test the effects of BA on the expression level of SULT1E1, a key enzyme in estrogen inactivation and elimination. RESULTS We found that TRF prevented HFD-induced embryo loss and alleviated the defect in luminal closure on D4.5 of pregnancy. The cyclic changes of E2 level were lost in mice fed ad libitum but not in TRF mice on the HFD. The HFD increased ERα expression and transcriptional activity, which induced AQP3 and AQP5 expression on D4.5 of pregnancy. TRF prevented the negative effect of the HFD on uterine luminal closure. Furthermore, in vitro and in vivo results showed that BA suppressed estrogen degradation by activating liver SULT1E1 expression. CONCLUSIONS Our findings demonstrated that TRF prevented HFD-induced defects in luminal closure, thereby improving embryonic implantation, and provide novel insights into the effects of dietary intervention on obesity and associated infertility.
Collapse
Affiliation(s)
- Luting Liu
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Yong Zhuo
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Haoqi Zhang
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Jing Li
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Xuemei Jiang
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Xingfa Han
- School of Life Sciences, Sichuan Agricultural University, Chengdu, P. R. China
| | - Jin Chao
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Bin Feng
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Lianqiang Che
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Shengyu Xu
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Yan Lin
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Jian Li
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Zhengfeng Fang
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130
| | - Mengmeng Sun
- College of Science, Sichuan Agricultural University, Xin Kang Road, Yucheng District, Ya'an 625014, P.R. China
| | - Ting Luo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - De Wu
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130.
| | - Lun Hua
- Animal Nutrition Institute, Chengdu, PR China, 611130; Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Chengdu, PR China, 611130; Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, PR China, 611130.
| |
Collapse
|
18
|
Harris C, Czaja K. Can Circadian Eating Pattern Adjustments Reduce Risk or Prevent Development of T2D? Nutrients 2023; 15:nu15071762. [PMID: 37049602 PMCID: PMC10096926 DOI: 10.3390/nu15071762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic condition that occurs in insulin-resistant people with reduced glucose uptake. It is contributed to and exacerbated by a poor diet that results in accumulation of adipose tissue, high blood sugar, and other metabolic issues. Because humans have undergone food scarcity throughout history, our species has adapted a fat reserve genotype. This adaptation is no longer beneficial, as eating at a higher frequency than that of our ancestors has had a significant effect on T2D development. Eating at high frequencies disrupts the circadian clock, the circadian rhythm, and the composition of the gut microbiome, as well as hormone secretion and sensitivity. The current literature suggests an improved diet requires meal consistency, avoiding late-night eating, low meal frequency, and fasting to increase metabolic health. In addition, fasting as a treatment for T2D must be used correctly for beneficial results. Early time-restricted eating (TRE) provides many benefits such as improving insulin resistance, cognitive function, and glycemic control. Alternate-day fasting (ADF), 5:2 fasting, and long-term fasting all have benefits; however, they may be less advantageous than early TRE. Therefore, eating pattern adjustments can be used to reduce T2D if used correctly.
Collapse
Affiliation(s)
- Carlee Harris
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Krzysztof Czaja
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
19
|
Aldamarany W, Taocui H, Liling D, Wanfu Y, Zhong G. Oral Supplementation with Three Vegetable Oils Differing in Fatty Acid Composition Alleviates High-Fat Diet-Induced Obesity in Mice by Regulating Inflammation and Lipid Metabolism. POL J FOOD NUTR SCI 2023. [DOI: 10.31883/pjfns/160186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
|
20
|
Ledesma-Pacheco SJ, Uriostegui-Pena AG, Rodriguez-Jacinto E, Gomez-Hernandez E, Estrada-Meza C, Banerjee A, Pathak S, Ruiz-Manriquez LM, Duttaroy AK, Paul S. Regulatory mechanisms of microRNAs in endocrine disorders and their therapeutic potential. Front Genet 2023; 14:1137017. [PMID: 36896239 PMCID: PMC9989203 DOI: 10.3389/fgene.2023.1137017] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/09/2023] [Indexed: 02/23/2023] Open
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding RNA molecules capable of regulating gene expression at the post-transcriptional level either by translational inhibition or mRNA degradation and have recently been importantly related to the diagnosis and prognosis of the most relevant endocrine disorders. The endocrine system comprises various highly vascularized ductless organs regulating metabolism, growth and development, and sexual function. Endocrine disorders constitute the fifth principal cause of death worldwide, and they are considered a significant public health problem due to their long-term effects and negative impact on the patient's quality of life. Over the last few years, miRNAs have been discovered to regulate various biological processes associated with endocrine disorders, which could be advantageous in developing new diagnostic and therapeutic tools. The present review aims to provide an overview of the most recent and significant information regarding the regulatory mechanism of miRNAs during the development of the most relevant endocrine disorders, including diabetes mellitus, thyroid diseases, osteoporosis, pituitary tumors, Cushing's syndrome, adrenal insufficiency and multiple endocrine neoplasia, and their potential implications as disease biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Luis M. Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Queretaro, Mexico
- Tecnologico de Monterrey, Escuela de Medicina, Monterrey, Mexico
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Queretaro, Mexico
| |
Collapse
|
21
|
Wang W, Liu Y, Li Y, Luo B, Lin Z, Chen K, Liu Y. Dietary patterns and cardiometabolic health: Clinical evidence and mechanism. MedComm (Beijing) 2023; 4:e212. [PMID: 36776765 PMCID: PMC9899878 DOI: 10.1002/mco2.212] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 02/08/2023] Open
Abstract
For centuries, the search for nutritional interventions to underpin cardiovascular treatment and prevention guidelines has contributed to the rapid development of the field of dietary patterns and cardiometabolic disease (CMD). Numerous studies have demonstrated that healthy dietary patterns with emphasis on food-based recommendations are the gold standard for extending lifespan and reducing the risks of CMD and mortality. Healthy dietary patterns include various permutations of energy restriction, macronutrients, and food intake patterns such as calorie restriction, intermittent fasting, Mediterranean diet, plant-based diets, etc. Early implementation of healthy dietary patterns in patients with CMD is encouraged, but an understanding of the mechanisms by which these patterns trigger cardiometabolic benefits remains incomplete. Hence, this review examined several dietary patterns that may improve cardiometabolic health, including restrictive dietary patterns, regional dietary patterns, and diets based on controlled macronutrients and food groups, summarizing cutting-edge evidence and potential mechanisms for CMD prevention and treatment. Particularly, considering individual differences in responses to dietary composition and nutritional changes in organ tissue diversity, we highlighted the critical role of individual gut microbiota in the crosstalk between diet and CMD and recommend a more precise and dynamic nutritional strategy for CMD by developing dietary patterns based on individual gut microbiota profiles.
Collapse
Affiliation(s)
- Wenting Wang
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yanfei Liu
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yiwen Li
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Binyu Luo
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Zhixiu Lin
- Faculty of MedicineThe Chinese University of Hong KongHong Kong
| | - Keji Chen
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Yue Liu
- National Clinical Research Centre for Chinese Medicine CardiologyXiyuan HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
22
|
Qi L, Chen Y. Circulating Bile Acids as Biomarkers for Disease Diagnosis and Prevention. J Clin Endocrinol Metab 2023; 108:251-270. [PMID: 36374935 DOI: 10.1210/clinem/dgac659] [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: 08/08/2022] [Revised: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022]
Abstract
CONTEXT Bile acids (BAs) are pivotal signaling molecules that regulate energy metabolism and inflammation. Recent epidemiological studies have reported specific alterations in circulating BA profiles in certain disease states, including obesity, type 2 diabetes mellitus (T2DM), nonalcoholic fatty liver disease (NAFLD), and Alzheimer disease (AD). In the past decade, breakthroughs have been made regarding the translation of BA profiling into clinical use for disease prediction. In this review, we summarize and synthesize recent data on variation in circulating BA profiles in patients with various diseases to evaluate the value of these biomarkers in human plasma for early diagnosis. EVIDENCE ACQUISITION This review is based on a collection of primary and review literature gathered from a PubMed search for BAs, obesity, T2DM, insulin resistance (IR), NAFLD, hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), colon cancer, and AD, among other keywords. EVIDENCE SYNTHESIS Individuals with obesity, T2DM, HCC, CCA, or AD showed specific alterations in circulating BA profiles. These alterations may have existed long before the initial diagnosis of these diseases. The intricate relationship between obesity, IR, and NAFLD complicates the establishment of clear and independent associations between BA profiles and nonalcoholic steatohepatitis. Alterations in the levels of total BAs and several BA species were seen across the entire spectrum of NAFLD, demonstrating significant increases with the worsening of histological features. CONCLUSIONS Aberrant circulating BA profiles are an early event in the onset and progression of obesity, T2DM, HCC, and AD. The pleiotropic effects of BAs explain these broad connections. Circulating BA profiles could provide a basis for the development of biomarkers for the diagnosis and prevention of a wide range of diseases.
Collapse
Affiliation(s)
- Li Qi
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
| | - Yongsheng Chen
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
23
|
Zhang W, Wang J, Wang L, Shi R, Chu C, Shi Z, Liu P, Li Y, Liu X, Liu Z. Alternate-day fasting prevents non-alcoholic fatty liver disease and working memory impairment in diet-induced obese mice. J Nutr Biochem 2022; 110:109146. [PMID: 36049672 DOI: 10.1016/j.jnutbio.2022.109146] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 04/28/2022] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Alternate-day fasting (ADF) regimen has been reported to alleviate obesity and insulin resistance. However, the effects of ADF on preventing diet-induced non-alcoholic fatty liver disease (NAFLD) and related cognitive deficits are still elusive. In the present study, a high-fat diet (HFD)-induced obese (DIO) C57BL/6 mouse model was established. Mice were treated with a 4-week long ADF regimen and/or switching the diet to a standard diet. ADF reduced lipid accumulation, activated AMPK/ULK1 signaling, and suppressed the phosphorylation of mTOR. Also, ADF inhibited lipid accumulation and inflammatory responses in the white adipose tissue and down-regulated expressions of PPAR-γ and cytokines. Moreover, ADF improved the working memory and synaptic structure in the DIO mice and upregulated PSD-95 and BDNF in the hippocampus. ADF reduced oxidative stress and microglial over-activation in the CNS. These results suggest that ADF attenuates NAFLD development in the liver of DIO mice, which is related to the mediating effects of ADF on autophagy and energy metabolism. ADF also enhanced cognitive function, which could be partly explained by the down-regulated peripheral inflammatory responses. This study indicates that ADF could be a promising intervention for alleviating NAFLD development and cognitive decline.
Collapse
Affiliation(s)
- Wentong Zhang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jun Wang
- Department of Digestive Diseases, Xijing Hospital, Xi'an, Shaanxi Province, China
| | - Luanfeng Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Renjie Shi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Chuanqi Chu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Zhiling Shi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Pujie Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Yitong Li
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province, China; Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
24
|
Mokhtari Z, Hosseini E, Hekmatdoost A, Haskey N, Gibson DL, Askari G. The effects of fasting diets on nonalcoholic fatty liver disease. Nutr Rev 2022:6809036. [DOI: 10.1093/nutrit/nuac092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease in the world. There is no confirmed treatment for NAFLD as yet. Recently, fasting regimens and their relationship to NAFLD have drawn a great deal of attention in the literature. We review the current evidence that supports fasting diets as an adjunctive therapeutic strategy for patients with NAFLD and address potential action mechanisms. We reason that the fasting diets might be a promising approach for modulating hepatic steatosis, fibroblast growth factors 19 and 21 signaling, lipophagy, and the metabolic profile.
Collapse
Affiliation(s)
- Zeinab Mokhtari
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences are with the , Isfahan, Iran
| | - Elham Hosseini
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences are with the , Isfahan, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and, Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences with the , Tehran, Iran
| | - Natasha Haskey
- Department of Biology, University of British Columbia—Okanagan Campus are with the , Kelowna, British Columbia, Canada
| | - Deanna L Gibson
- Department of Biology, University of British Columbia—Okanagan Campus are with the , Kelowna, British Columbia, Canada
| | - Gholamreza Askari
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences are with the , Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences with the , Isfahan, Iran
| |
Collapse
|
25
|
Zhou J, Jiang Z, Lin Y, Li C, Liu J, Tian M, Liu Y, Chen K. The daily caloric restriction and alternate-day fasting ameliorated lipid dysregulation in type 2 diabetic mice by downregulating hepatic pescadillo 1. Eur J Nutr 2022; 61:2775-2797. [PMID: 35290477 DOI: 10.1007/s00394-022-02850-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/22/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE A possible link between pescadillo 1 (PES1) and lipid metabolism has been reported. However, whether PES1 is involved in the effects of daily caloric restriction (CR) and alternate-day fasting (ADF) interventions on diabetes-related lipid dysregulation is not elucidated. The current study aims are to explore the role of PES1 in effects of CR and ADF on diabetic mice and related mechanism. METHODS Eight-week-old male db/db mice with type 2 diabetes mellitus (T2DM) were randomly divided into untreated T2DM, CR and ADF groups. McArdle hepatocytes were treated with 48 h high glucose (HG), 48 h normal glucose (NG) and 24 h HG plus 24 h NG, respectively. Pes1 siRNA and overexpression plasmid were, respectively, transfected into liver cells, and AAV9-Pes1-shRNA was injected into db/db mice. RESULTS After 12-week interventions, the peroxisome proliferator-activated receptor alpha (PPAR-α) and carnitine palmitoyltransferase 1A (CPT1A) levels in livers of T2DM mice were enhanced by CR and ADF interventions with reductions of hepatic and plasma triglycerides. Unexpectedly, hepatic PES1 levels were downregulated by two interventions, consistent with the results of 48 h NG and 24 h HG plus 24 h NG-treated cells. Moreover, CPT1A level was upregulated in Pes1-siRNA-treated cells and AAV9-Pes1-shRNA injected murine livers, in contrast to Pes1 overexpression in cultured cells. Mechanistically, 48 h NG or 24 h HG plus 24 h NG treatment increased PPAR-α binding to Pes1 promoter, suppressing the PES1 expression, thereby lowering the PES1-mediated ubiquitination of CPT1A. CONCLUSION The present study suggests that CR and ADF may improve lipid dysregulation in diabetic mice by downregulating hepatic PES1.
Collapse
Affiliation(s)
- Jielin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Zhengxuan Jiang
- Department of Ophthalmology, The Second Affiliated Hospital, Anhui Medical University, Hefei, 230021, Anhui, China
| | - Yan Lin
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Chengcheng Li
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Juan Liu
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Mengjun Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yong Liu
- AIER Hefei Eye Hospital Affiliated To Anhui Medical University, Hefei, 230031, Anhui, China.
| | - Keyang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
26
|
Cui J, Lee S, Sun Y, Zhang C, Hill MA, Li Y, Zhang H. Alternate Day Fasting Improves Endothelial Function in Type 2 Diabetic Mice: Role of Adipose-Derived Hormones. Front Cardiovasc Med 2022; 9:925080. [PMID: 35711339 PMCID: PMC9196729 DOI: 10.3389/fcvm.2022.925080] [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] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Intermittent fasting, including alternate day fasting (ADF), has grown in popularity as it can produce clinically significant metabolic benefits and is often considered to be easier to adhere to than other types of diets such as chronic calorie restriction. However, the effects of ADF on diabetes-associated vascular dysfunction, and the role of adipose-derived hormones, i.e., adipokines, in mediating its effects, remain largely unknown. Objective We aimed to test the hypothesis that ADF protects against diabetes-associated endothelial dysfunction, at least partly through modulating adipokine profiles. Methods Control mice (m Lepr db ) and diabetic mice (Leprdb ) were treated with 12-weeks of ADF. Glucose metabolism, endothelial function, and adipokine profile were assessed. Results ADF reduced fasting blood glucose level and homeostatic model assessment for insulin resistance (HOMA-IR), and improved insulin sensitivity. ADF improved endothelium-dependent vasorelaxation of small mesenteric arteries (SMA) of Leprdb mice. The improvement in endothelial function was largely attenuated by incubation with the nitric oxide synthase inhibitor, L-NAME. These ADF-induced metabolic and vascular benefits were accompanied by increased circulating adiponectin. Adenovirus-mediated adiponectin supplementation improved endothelial function in Leprdb mice, supporting endothelial protective roles in diabetes-associated endothelial dysfunction. Protein tyrosine nitration is a post-translational modification that serves as a marker of oxidative stress. Nitrotyrosine protein levels in SMA and mesenteric adipose tissue (MAT) were elevated in Leprdb mice. ADF reduced nitrotyrosine protein in SMA, but not in MAT, of Leprdb mice. Conclusion ADF exerts metabolic and endothelial protective benefits. The improvement of endothelial function was partly mediated by increased adiponectin, representing an important mechanism for the beneficial vascular effects resulting from ADF.
Collapse
Affiliation(s)
- Jian Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sewon Lee
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
- Division of Sport Science, College of Arts and Physical Education, Incheon National University, Incheon, South Korea
| | - Yan Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cuihua Zhang
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Yuhang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hanrui Zhang
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
- Department of Medicine, Division of Cardiology, Cardiometabolic Genomics Program, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
27
|
Wang X, Lv W, Xu J, Zheng A, Zeng M, Cao K, Wang X, Cui Y, Li H, Yang M, Shao Y, Zhang F, Zou X, Long J, Feng Z, Liu J. Hepatic Suppression of Mitochondrial Complex II Assembly Drives Systemic Metabolic Benefits. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105587. [PMID: 35037426 PMCID: PMC8948583 DOI: 10.1002/advs.202105587] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Indexed: 05/05/2023]
Abstract
Alternate day fasting (ADF), the most popular form of caloric restriction, has shown to improve metabolic health in preclinical subjects, while intrinsic network underpinning the process remains unclear. Here, it is found that liver undergoes dramatic metabolic reprogramming during ADF, accompanied surprisingly with unique complex II dysfunction attributing to suspended complex II assembly via suppressing SDHAF4, a recently identified assembly factor. Despite moderate mitochondrial complex II dysfunction, hepatic Sdhaf4 knockout mice present intriguingly improved glucose tolerance and systemic insulin sensitivity, consistent with mice after ADF intervention. Mechanistically, it is found that hepatocytes activate arginine-nitric oxide (NO) biosynthesis axle in response to complex II and citric acid cycle dysfunction, the release of NO from liver can target muscle and adipocytes in addition to its autocrine action for enhanced insulin sensitivity. These results highlight the pivotal role of liver in ADF-associated systemic benefits, and suggest that targeting hepatic complex II assembly can be an intriguing strategy against metabolic disorders.
Collapse
Affiliation(s)
- Xueqiang Wang
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Jie Xu
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Adi Zheng
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Mengqi Zeng
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Ke Cao
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Xun Wang
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Yuting Cui
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Hao Li
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Meng Yang
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Yongping Shao
- Frontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Fang Zhang
- Department of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineXi'anShanghai200240China
- National Clinical Research Center for Eye DiseasesShanghai200240China
| | - Xuan Zou
- National & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShannxi710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShannxi710004China
| | - Jiangang Long
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zhihui Feng
- Frontier Institute of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- National & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShannxi710004China
- University of Health and Rehabilitation SciencesQingdaoShandong266071China
| | - Jiankang Liu
- Center for Mitochondrial Biology and MedicineThe Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- University of Health and Rehabilitation SciencesQingdaoShandong266071China
| |
Collapse
|
28
|
Zhao Y, Jia M, Chen W, Liu Z. The neuroprotective effects of intermittent fasting on brain aging and neurodegenerative diseases via regulating mitochondrial function. Free Radic Biol Med 2022; 182:206-218. [PMID: 35218914 DOI: 10.1016/j.freeradbiomed.2022.02.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/29/2022] [Accepted: 02/21/2022] [Indexed: 12/11/2022]
Abstract
Intermittent fasting (IF) has been studied for its effects on lifespan and the prevention or delay of age-related diseases upon the regulation of metabolic pathways. Mitochondria participate in key metabolic pathways and play important roles in maintaining intracellular signaling networks that modulate various cellular functions. Mitochondrial dysfunction has been described as an early feature of brain aging and neurodegeneration. Although IF has been shown to prevent brain aging and neurodegeneration, the mechanism is still unclear. This review focuses on the mechanisms by which IF improves mitochondrial function, which plays a central role in brain aging and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. The cellular and molecular mechanisms of IF in brain aging and neurodegeneration involve activation of adaptive cellular stress responses and signaling- and transcriptional pathways, thereby enhancing mitochondrial function, by promoting energy metabolism and reducing oxidant production.
Collapse
Affiliation(s)
- Yihang Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Mengzhen Jia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Weixuan Chen
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China; German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
| |
Collapse
|
29
|
Liu X, Zhang Y, Ma C, Lin J, Du J. Alternate-day fasting alleviates high fat diet induced non-alcoholic fatty liver disease through controlling PPARα/Fgf21 signaling. Mol Biol Rep 2022; 49:3113-3122. [PMID: 35107741 DOI: 10.1007/s11033-022-07142-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/11/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease that ultimately leads to cirrhosis and hepatocellular carcinoma. Intermittent fasting has been proposed as a nonpharmacological dietary approach against metabolic diseases, including NAFLD. In this study, we aimed to investigate the effect of alternate day fasting (ADF) on high-fat diet (HFD)-induced NAFLD in C57BL/6 mice. METHODS A mouse model of fatty liver disease was established by feeding the mice a HFD for 16 weeks. The mice were administered by body weight, lipid accumulation and inflammation. PPARα, FGF21, serum triglycerides (TG), total cholesterol (TC), transaminase and lipogenesis were assessed. RESULTS The results showed that long-term ADF can attenuate fatty liver disease by reducing hepatic inflammation and lipid accumulation in a mouse model. Meanwhile, fasting elevated the expression of serum and hepatic fibroblast growth Factor 21 (Fgf21), a circulating hormone produced predominantly in the liver, and could effectively prevent and ameliorate the pathogenesis of NAFLD. Serum starvation also enhanced Fgf21 expression and reduced free fatty acid (FFA)-induced lipid storage in hepatocytes. Moreover, refeeding inhibited the increase in Fgf21 expression induced by fasting. This fasted-to-refed transition is closely related to the expression of Fgf21. Further in vitro and in vivo studies showed that fasting-sensitive PPARα is indispensable for the expression of Fgf21 and its protective effect on NAFLD. CONCLUSION These findings indicated that long-term ADF protects mouse livers against HFD induced fatty liver disease through controlling PPARα/Fgf21 signaling. In conclusion, ADF can emerge as a non-pharmacological dietary approach against fatty liver disease.
Collapse
Affiliation(s)
- Xinlei Liu
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yan Zhang
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chunya Ma
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China.,Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China. .,Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Jiang Du
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Medical Engineering, Xinxiang Medical University, East of JinSui Road #601, Xinxiang City, 453003, Henan Province, China. .,Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
30
|
Raji-Amirhasani A, Khaksari M, Shahrokhi N, Soltani Z, Nazari-Robati M, Mahani FD, Hajializadeh Z, Sabet N. Comparison of the effects of different dietary regimens on susceptibility to experimental acute kidney injury: the role of SIRT1 and TGF-β1. Nutrition 2022; 96:111588. [DOI: 10.1016/j.nut.2022.111588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/10/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
|
31
|
Zhong H, Wang J, Abdullah, Hafeez MA, Guan R, Feng F. Lactobacillus plantarum ZJUFB2 Prevents High Fat Diet-Induced Insulin Resistance in Association With Modulation of the Gut Microbiota. Front Nutr 2021; 8:754222. [PMID: 34805244 PMCID: PMC8604096 DOI: 10.3389/fnut.2021.754222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lactobacillus plantarum ZJUFB2 is a novel probiotic isolate derived from Chinese traditional sourdough that possesses promising probiotics properties. This study aimed to investigate the effects of L. plantarum ZJUFB2 (B2) on insulin sensitivity using mice fed on a high-fat diet (HFD) as well as to explore the involved mechanisms. Purposely, male C57BL/6 mice continuously received an intervention of B2 (~109 CFU/day) for 16 weeks. The results showed that B2 treatment remarkably ameliorated insulin resistance and hyperglycemia in HFD-fed mice. The B2 intervention significantly decreased the hepatic lipid accumulation, serum low-density lipoproteins cholesterol, and lipopolysaccharides, and regulated the bile acids levels as well as liver mRNA expression involved in lipid metabolism. Moreover, the B2 intervention significantly changed the gut microbiota, specifically, showed a lower abundance of obesity-related and inflammation-associated microbes, e.g., Ruminococcus and Mogibacteriaceae. Furthermore, it exhibited a higher abundance of short-chain fatty acids and bile salt hydrolas-producing bacteria, such as Bifidobacterium and F16 compared with the HFD group. The findings of this study suggested that B2 is a novel probiotic, effective in preventing insulin resistance by improving the gut microbiota and bile acids.
Collapse
Affiliation(s)
- Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Jian Wang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Abdullah
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, China
| | - Muhammad Adnan Hafeez
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Liu Q, Xie T, Xi Y, Li L, Mo F, Liu X, Liu Z, Gao JM, Yuan T. Sesamol Attenuates Amyloid Peptide Accumulation and Cognitive Deficits in APP/PS1 Mice: The Mediating Role of the Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12717-12729. [PMID: 34669408 DOI: 10.1021/acs.jafc.1c04687] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Alzheimer's disease (AD), a neurodegenerative disease, is the leading cause of dementia. Sesamol is a lignan extracted from sesame oil and has been found to exert neuroprotective effects. The present study aimed to investigate the neuroprotective effects of sesamol on APPswe/PS1dE9 transgenic AD mice. The AD mice were fed with a diet supplemented with sesamol (0.075 w/w %). Sesamol treatment improved spatial memory and learning ability in AD mice, improved neuronal damage, and decreased Aβ accumulation. Sesamol protected the synaptic ultrastructure and inhibited neuroinflammatory responses in the brain of AD mice. Sesamol also significantly inhibited the overactivated microglia and reduced the overexpression of TNF-α and IL-1β in the brain of AD mice. Notably, sesamol reshaped gut microbiota by significantly decreasing the relative abundance of Helicobacter hepaticus, Clostridium, and Bacillaceae, enhancing the relative abundance of Rikenellaceae and Bifidobacterium in AD mice. It has been found that sesamol protected the gut barrier integrity and prevented the LPS leakage into the serum. Importantly, sesamol remarkably enhanced the content of SCFAs, including acetate, propionate, isobutyrate, butyrate, and valerate, in AD mice. Correlation analysis indicated that there was a strong correlation between the levels of SCFAs and cognitive functions. These results demonstrated that sesamol attenuated AD-related cognitive dysfunction and neuroinflammatory responses, which could be partly explained by its role in mediating the gut microbe-SCFA-brain axis. Thus, sesamol is a promising nutritional intervention strategy to prevent AD via the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Qing Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Tianzhi Xie
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yujia Xi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ling Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fengfeng Mo
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
- Department of Food Science, Cornell University, Ithaca, New York 14853, United States
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, P. R. China
| | - Tian Yuan
- Department of Food Science, Cornell University, Ithaca, New York 14853, United States
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, P. R. China
| |
Collapse
|
33
|
Yan H, Wei W, Hu L, Zhang Y, Zhang H, Liu J. Reduced Feeding Frequency Improves Feed Efficiency Associated With Altered Fecal Microbiota and Bile Acid Composition in Pigs. Front Microbiol 2021; 12:761210. [PMID: 34712219 PMCID: PMC8546368 DOI: 10.3389/fmicb.2021.761210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
A biphasic feeding regimen exerts an improvement effect on feed efficiency of pigs. While gut microbiome and metabolome are known to affect the host phenotype, so far the effects of reduced feeding frequency on fecal microbiota and their metabolism in pigs remain unclear. Here, the combination of 16S rRNA sequencing technique as well as untargeted and targeted metabolome analyses was adopted to investigate the fecal microbiome and metabolome of growing–finishing pigs in response to a biphasic feeding [two meals per day (M2)] pattern. Sixty crossbred barrows were randomly assigned into two groups with 10 replicates (three pigs/pen), namely, the free-access feeding group (FA) and the M2 group. Pigs in the FA group were fed free access while those in the M2 group were fed ad libitum twice daily for 1 h at 8:00 and 18:00. Results showed that pigs fed biphasically exhibited increased feed efficiency compared to FA pigs. The Shannon and Simpson indexes were significantly increased by reducing the feeding frequency. In the biphasic-fed pigs, the relative abundances of Subdoligranulum, Roseburia, Mitsuokella, and Terrisporobacter were significantly increased while the relative abundances of unidentified_Spirochaetaceae, Methanobrevibacter, unidentified_Bacteroidales, Alloprevotella, Parabacteroides, and Bacteroides were significantly decreased compared to FA pigs. Partial least-square discriminant analysis (PLS-DA) analysis revealed an obvious variation between the FA and M2 groups; the differential features were mainly involved in arginine, proline, glycine, serine, threonine, and tryptophan metabolism as well as primary bile acid (BA) biosynthesis. In addition, the changes in the microbial genera were correlated with the differential fecal metabolites. A biphasic feeding regimen significantly increased the abundances of primary BAs and secondary BAs in feces of pigs, and the differentially enriched BAs were positively correlated with some specific genera. Taken together, these results suggest that the improvement effect of a reduced feeding frequency on feed efficiency of pigs might be associated with the altered fecal microbial composition and fecal metabolite profile in particular the enlarged stool BA pool.
Collapse
Affiliation(s)
- Honglin Yan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Wenzhuo Wei
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Luga Hu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Yong Zhang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| |
Collapse
|
34
|
Qi L, Tian Y, Chen Y. Circulating Bile Acid Profiles: A Need for Further Examination. J Clin Endocrinol Metab 2021; 106:3093-3112. [PMID: 34279029 DOI: 10.1210/clinem/dgab531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Bile acids (BAs) are increasingly recognized as metabolic and chronobiologic integrators that synchronize the systemic metabolic response to nutrient availability. Alterations in the concentration and/or composition of circulating BAs are associated with a number of metabolic disorders, such as obesity, type 2 diabetes mellitus (T2DM), insulin resistance (IR), and metabolic associated fatty liver disease (MAFLD). This review summarizes recent evidence that links abnormal circulating BA profiles to multiple metabolic disorders, and discusses the possible mechanisms underlying the connections to determine the role of BA profiling as a novel biomarker for these abnormalities. EVIDENCE ACQUISITION The review is based on a collection of primary and review literature gathered from a PubMed search of BAs, T2DM, IR, and MAFLD, among other keywords. EVIDENCE SYNTHESIS Obese and IR subjects appear to have elevated fasting circulating BAs but lower postprandial increase when compared with controls. The possible underlying mechanisms are disruption in the synchronization between the feeding/fasting cycle and the properties of BA-regulated metabolic pathways. Whether BA alterations are associated per se with MAFLD remains inconclusive. However, increased fasting circulating BAs level was associated with higher risk of advanced fibrosis stage. Thus, for patients with MAFLD, dynamically monitoring the circulating BA profiles may be a promising tool for the stratification of MAFLD. CONCLUSIONS Alterations in the concentration, composition, and rhythm of circulating BAs are associated with adverse events in systemic metabolism. Subsequent investigations regarding these aspects of circulating BA kinetics may help predict future metabolic disorders and guide therapeutic interventions.
Collapse
Affiliation(s)
- Li Qi
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yongsheng Chen
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| |
Collapse
|
35
|
Abstract
Fasting potentials are the most interesting topics in the Nutritional Era. Fasting consists of the catabolism of lipids, proteins, and carbohydrates to maintain blood glucose levels in a normal range. The action mechanisms of fasting were firstly understood in minor organisms and later in humans. Nutritional interventions of caloric restriction could attenuate age-associated epigenetic alterations and could have a protective effect against cellular alterations, promoting longevity and health span. While most fasting studies point out the weight and fat mass decreases, it is important to define specific guidelines for fasting and non-fasting days to enhance adherence, minimize the dropout rates of the interventions, and maximize body composition improvement. Although the panorama of evidence on fasting and caloric restriction is wide, there is a lack of a safe fasting protocol to guide physicians in its prescription. The main goal is to identify a how to use guide, a major posology of fasting, inserted within a huge dietetic personalized strategy leading to an optimal and healthy nutritional status.
Collapse
|
36
|
Shi H, Zhang B, Abo-Hamzy T, Nelson JW, Ambati CSR, Petrosino JF, Bryan RM, Durgan DJ. Restructuring the Gut Microbiota by Intermittent Fasting Lowers Blood Pressure. Circ Res 2021; 128:1240-1254. [PMID: 33596669 PMCID: PMC8085162 DOI: 10.1161/circresaha.120.318155] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/17/2021] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Huanan Shi
- Department of Anesthesiology, Baylor College of Medicine,
Houston TX, USA
- Department of Molecular Physiology and Biophysics, Baylor
College of Medicine, Houston TX, USA
| | - Bojun Zhang
- Department of Anesthesiology, Baylor College of Medicine,
Houston TX, USA
| | - Taylor Abo-Hamzy
- School of Health Professions, Baylor College of Medicine,
Houston TX, USA
| | - James W. Nelson
- Department of Anesthesiology, Baylor College of Medicine,
Houston TX, USA
- Integrated Molecular and Biomedical Sciences Graduate
Program, Baylor College of Medicine, Houston TX, USA
| | | | - Joseph F. Petrosino
- Department of Molecular Virology and Microbiology, The
Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine,
Houston TX, USA
| | - Robert M. Bryan
- Department of Anesthesiology, Baylor College of Medicine,
Houston TX, USA
- Department of Molecular Physiology and Biophysics, Baylor
College of Medicine, Houston TX, USA
| | - David J. Durgan
- Department of Anesthesiology, Baylor College of Medicine,
Houston TX, USA
- Department of Molecular Physiology and Biophysics, Baylor
College of Medicine, Houston TX, USA
| |
Collapse
|
37
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
38
|
Zou Q, Zhang X, Liu X, Li Y, Tan Q, Dan Q, Yuan T, Liu X, Liu RH, Liu Z. Ficus carica polysaccharide attenuates DSS-induced ulcerative colitis in C57BL/6 mice. Food Funct 2020; 11:6666-6679. [PMID: 32658237 DOI: 10.1039/d0fo01162b] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Ficus carica polysaccharide (FCPS) components of the common fig fruit have been demonstrated to exhibit antioxidant and immunity-enhancing activities. However, it is unclear whether it could prevent the ulcerative colitis development. Here, we reported that 5 week orally administered FCPS (150-300 mg per kg bw) significantly prevented DSS-induced colitis in C57BL/6J mice by improving the colon length and suppressing the infiltration of inflammatory cells in the gut. FCPS treatment protected the goblet cells, elevated the expression of tight junction protein claudin-1, and suppressed the formation of cytokines including TNF-α and IL-1β. FCPS supplementation significantly reformed the gut microbiome by enhancing the abundance of S24-7, Bacteroides, and Coprococus, and suppressing the abundance of Escherichia and Clostridium at the genus level. Consistently, the formation of beneficial microbial metabolites, short chain fatty acids, especially acetate and butyrate, were improved in FCPS-treated colitis mice. The correlation analysis indicated that the protective effects of FCPS on ulcerative colitis might be highly correlated with the microbiota composition changes and the formation of SCFAs. In conclusion, these results indicated that FCPS supplementation could be a promising nutritional strategy for reducing inflammatory bowel disease and the gut microbes play essential roles in providing these beneficial effects.
Collapse
Affiliation(s)
- Qianhui Zou
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Mulati A, Ma S, Zhang H, Ren B, Zhao B, Wang L, Liu X, Zhao T, Kamanova S, Sair AT, Liu Z, Liu X. Sea-Buckthorn Flavonoids Alleviate High-Fat and High-Fructose Diet-Induced Cognitive Impairment by Inhibiting Insulin Resistance and Neuroinflammation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:5835-5846. [PMID: 32363873 DOI: 10.1021/acs.jafc.0c00876] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Sea-buckthorn flavonoids (SFs) have been used as functional food components for their bioactive potential in preventing metabolic complications caused by diet, such as obesity and inflammation. However, the protective effect of SFs on cognitive functions is not fully clear. In this study, a high-fat and high-fructose diet (HFFD)-induced obese mice model was treated with SFs for 14 weeks. It was found that the oral SF administration (0.06% and 0.31% w/w, mixed in diet) significantly reduced bodyweight gain and insulin resistance in the HFFD-fed mice. SFs significantly prevented HFFD-induced neuronal loss and memory impairment in behavioral tests. Additionally, SFs also suppressed the HFFD-induced synaptic dysfunction and neuronal damages by increasing the protein expressions of PSD-95. Furthermore, SF treatment activated the ERK/CREB/BDNF and IRS-1/AKT pathways and inactivated the NF-κB signaling and its downstream inflammatory mediator expressions. In conclusion, SFs are a potential nutraceutical to prevent high-energy density diet-induced cognitive impairments, which could be possibly explained by their mediating effects on insulin signaling and inflammatory responses in the brain.
Collapse
Affiliation(s)
- Aiziguli Mulati
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Shaobo Ma
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Hongbo Zhang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Bo Ren
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Beita Zhao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Luanfeng Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Xiaoning Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Tong Zhao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Svetlana Kamanova
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| | - Ali Tahir Sair
- Department of Food Science, Cornell University, Ithaca, New York 14850, United States
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
- Department of Food Science, Cornell University, Ithaca, New York 14850, United States
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 China
| |
Collapse
|