1
|
Liu T, Ji X, Zang H, Li Z, Yao W, Wan L, Zhang C, Zhang Y. Endoplasmic reticulum stress: The underlying mechanism of chronic pain. Neurobiol Dis 2024; 202:106697. [PMID: 39389155 DOI: 10.1016/j.nbd.2024.106697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
Chronic pain (CP) affects over 30 % of the global population, imposing significant financial burdens on individuals and society. However, existing treatments for CP offer limited efficacy and troublesome side effects, primarily owing to a lack of knowledge of its precise underlying mechanism. Pathological stimuli disrupt the intricate process of protein folding and endoplasmic reticulum (ER) homeostasis. This disruption leads to the accumulation of misfolded or unfolded proteins in the ER, generating a condition termed ER stress. Emerging data have indicated that ER stress, occurring in the peripheral and central nervous systems, contributes to the development and maintenance of CP. This review aimed to comprehensively explore the intersection of ER stress and CP within the lower and upper nervous systems and highlight the cell-specific contributions of the unfolded protein response in different CP types. We provide a comprehensive synthesis of evidence from animal models, examining neuronal and non-neuronal mechanisms and discuss the damaging ER stress-linked inflammation, autophagy, oxidative stress, and apoptosis, which collectively drive disease progression and contribute to a neurotoxic environment. However, the mechanisms through which ER stress influences the most advanced centre-of-pain projections in the brain remain unclear. Further investigation in this area is crucial to elucidate the relationship between ER stress and CP and facilitate the development of novel therapeutic drugs for this intractable dilemma.
Collapse
Affiliation(s)
- Tongtong Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Ji
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Zang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zuofan Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenlong Yao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Wan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanhan Zhang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Zhang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Liu Q, Wang M, Hou Y, Chen R, Liu H, Han T, Liu D. Deciphering the multifaceted effects of artificial sweeteners on body health and metabolic functions: a comprehensive review and future perspectives. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 39368060 DOI: 10.1080/10408398.2024.2411410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
As the rates of chronic diseases such as obesity and diabetes rise worldwide, there is a growing demand for low-calorie or no-calorie sweeteners to reduce sugar intake without sacrificing the sweetness of foods and beverages. Artificial sweeteners have become indispensable as substitutes for sugar due to their high sweetening power and low impact on blood sugar levels and are used in a variety of low-calorie foods and beverages. Although artificial sweeteners offer an alternative for reducing sugar intake while maintaining sweetness, research into their long-term health effects, particularly at high doses, is ongoing, further scientific research and regulatory review are needed to clarify these potential health risks. This article reviews the latest research on the health effects of artificial sweeteners, based on recent studies, introduces the classification, performance, and safety standards for artificial sweeteners, analyses their potential harms to the nervous, immune, and circulatory systems, reproductive system, as well as their effects on gut microbiota, liver function, cancer, diabetes, and obesity. In addition, consumer perceptions of artificial sweeteners and future research directions are discussed, providing insights into current research controversies and knowledge gaps, as well as the health research and market application of artificial sweeteners.
Collapse
Affiliation(s)
- Qiang Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Min Wang
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Yuting Hou
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Meat Innovation Center of Liaoning Province, Jinzhou, China
- Liaoning Kazuo Hybrid Wild Boar Science and Technology Backyard, Chaoyang, China
| | - Rui Chen
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Haixia Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Tianlong Han
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Liaoning Kazuo Hybrid Wild Boar Science and Technology Backyard, Chaoyang, China
| | - Dengyong Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Meat Innovation Center of Liaoning Province, Jinzhou, China
| |
Collapse
|
3
|
Mao H, Kim GH, Pan L, Qi L. Regulation of leptin signaling and diet-induced obesity by SEL1L-HRD1 ER-associated degradation in POMC expressing neurons. Nat Commun 2024; 15:8435. [PMID: 39343970 PMCID: PMC11439921 DOI: 10.1038/s41467-024-52743-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
Endoplasmic reticulum (ER) homeostasis in the hypothalamus has been implicated in the pathogenesis of diet-induced obesity (DIO) and type 2 diabetes; however, the underlying molecular mechanism remain vague and debatable. Here we report that SEL1L-HRD1 protein complex of the highly conserved ER-associated protein degradation (ERAD) machinery in POMC-expressing neurons ameliorates diet-induced obesity and its associated complications, partly by regulating the turnover of the long isoform of Leptin receptors (LepRb). Loss of SEL1L in POMC-expressing neurons attenuates leptin signaling and predisposes mice to HFD-associated pathologies including fatty liver, glucose intolerance, insulin and leptin resistance. Mechanistically, nascent LepRb, both wildtype and disease-associated Cys604Ser variant, are misfolding prone and bona fide substrates of SEL1L-HRD1 ERAD. In the absence of SEL1L-HRD1 ERAD, LepRb are largely retained in the ER, in an ER stress-independent manner. This study uncovers an important role of SEL1L-HRD1 ERAD in the pathogenesis of central leptin resistance and leptin signaling.
Collapse
Affiliation(s)
- Hancheng Mao
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Geun Hyang Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York, NY, 10591, USA
| | - Linxiu Pan
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Ling Qi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
4
|
Lun W, Yan Q, Guo X, Zhou M, Bai Y, He J, Cao H, Che Q, Guo J, Su Z. Mechanism of action of the bile acid receptor TGR5 in obesity. Acta Pharm Sin B 2024; 14:468-491. [PMID: 38322325 PMCID: PMC10840437 DOI: 10.1016/j.apsb.2023.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/17/2023] [Accepted: 10/24/2023] [Indexed: 02/08/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of membrane protein receptors, and Takeda G protein-coupled receptor 5 (TGR5) is a member of this family. As a membrane receptor, TGR5 is widely distributed in different parts of the human body and plays a vital role in regulating metabolism, including the processes of energy consumption, weight loss and blood glucose homeostasis. Recent studies have shown that TGR5 plays an important role in glucose and lipid metabolism disorders such as fatty liver, obesity and diabetes. With the global obesity situation becoming more and more serious, a comprehensive explanation of the mechanism of TGR5 and filling the gaps in knowledge concerning clinical ligand drugs are urgently needed. In this review, we mainly explain the anti-obesity mechanism of TGR5 to promote the further study of this target, and show the electron microscope structure of TGR5 and review recent studies on TGR5 ligands to illustrate the specific binding between TGR5 receptor binding sites and ligands, which can effectively provide new ideas for ligand research and promote drug research.
Collapse
Affiliation(s)
- Weijun Lun
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qihao Yan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xinghua Guo
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Minchuan Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Jincan He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Science City, Guangzhou 510663, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
5
|
Kim HR, Young CN. Circumventricular organ-hypothalamic circuit endoplasmic reticulum stress drives hepatic steatosis during obesity. Obesity (Silver Spring) 2024; 32:59-69. [PMID: 37794528 DOI: 10.1002/oby.23895] [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: 06/09/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD), characterized by excess liver triglyceride accumulation (hepatic steatosis), leads to an increased risk for cardiometabolic diseases and obesity-related mortality. Emerging evidence points to endoplasmic reticulum (ER) stress in the central nervous system as critical in NAFLD pathogenesis. Here, we tested the contribution of ER stress in a circumventricular organ-hypothalamic circuit in NAFLD development during obesity. METHODS C57BL/6J male mice were fed a high-fat diet (HFD) or normal chow. A combination of histological, viral tracing, intersectional viral targeting, and in vivo integrative physiological approaches were used to examine the role of ER stress in subfornical organ to hypothalamic paraventricular nucleus projecting neurons (SFO➔PVN) in NAFLD during diet-induced obesity. RESULTS Immunohistochemical analysis revealed marked unfolded protein response activation in the SFO, particularly in excitatory SFO➔PVN neurons of HFD-fed animals. Moreover, intersectional viral inhibition of ER stress in SFO➔PVN neurons resulted in a reduction in hepatomegaly, hepatic steatosis, and a blunted increase in body weight gain during diet-induced obesity, independent of changes in food intake, substrate partitioning, energy expenditure, and ambulatory activity. CONCLUSIONS These results indicate that ER stress in an SFO➔PVN neural circuit contributes to hepatic steatosis during obesity.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Colin N Young
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
6
|
Abasubong KP, Jiang GZ, Guo HX, Wang X, Li XF, Yan-Zou D, Liu WB, Desouky HE. High-fat diet alters intestinal microbiota and induces endoplasmic reticulum stress via the activation of apoptosis and inflammation in blunt snout bream. FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:1079-1095. [PMID: 37831370 DOI: 10.1007/s10695-023-01240-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/16/2023] [Indexed: 10/14/2023]
Abstract
The primary organ for absorbing dietary fat is the gut. High dietary lipid intake negatively affects health and absorption by causing fat deposition in the intestine. This research explores the effect of a high-fat diet (HFD) on intestinal microbiota and its connections with endoplasmic reticulum stress and inflammation. 60 fish (average weight: 45.84 ± 0.07 g) were randomly fed a control diet (6% fat) and a high-fat diet (12 % fat) in four replicates for 12 weeks. From the result, hepatosomatic index (HSI), Visceralsomatic index (VSI), abdominal fat (ADF), Intestosomatic index (ISI), mesenteric fat (MFI), Triglycerides (TG), total cholesterol (TC), non-esterified fatty acid (NEFA) content were substantially greater on HFD compared to the control diet. Moreover, fish provided the HFD significantly obtained lower superoxide dismutase (SOD) and glutathione peroxidase (GPX) activities. In contrast, an opposite result was seen in malondialdehyde (MDA) content in comparison to the control. HFD significantly altered intestinal microbiota in blunt snout bream, characterized by an increased abundance of Aeromonas, Plesiomonas proteobacteria, and firmicutes with a reduced abundance of Cetobacterium and ZOR0006. The transcriptional levels of glucose-regulated protein 78 (grp78), inositol requiring enzyme 1 (ire1), spliced X box-binding protein 1 (xbp1), DnaJ heat shock protein family (Hsp40) member B9 (dnajb9), tumor necrosis factor alpha (tnf-α), nuclear factor-kappa B (nf-κb), monocyte chemoattractant protein-1 (mcp-1), and interleukin-6 (il-6) in the intestine were markedly upregulated in fish fed HFD than the control group. Also, the outcome was similar in bax, caspases-3, and caspases-9, ZO-1, Occludin-1, and Occludin-2 expressions. In conclusion, HFD could alter microbiota and facilitate chronic inflammatory signals via activating endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Kenneth Prudence Abasubong
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Guang-Zhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Hui-Xing Guo
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Xi Wang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Xiang-Fei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Dong Yan-Zou
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Wen-Bin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China.
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China.
| | - Hesham Eed Desouky
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
- Department of Animal and Poultry Production, Faculty of Agriculture, Damanhour University, Damanhour, Beheria, 22713, Egypt
| |
Collapse
|
7
|
Zhou M, Huang J, Zhou J, Zhi C, Bai Y, Che Q, Cao H, Guo J, Su Z. Anti-Obesity Effect and Mechanism of Chitooligosaccharides Were Revealed Based on Lipidomics in Diet-Induced Obese Mice. Molecules 2023; 28:5595. [PMID: 37513467 PMCID: PMC10384603 DOI: 10.3390/molecules28145595] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Chitooligosaccharide (COS) is a natural product from the ocean, and while many studies have reported its important role in metabolic diseases, no study has systematically elaborated the anti-obesity effect and mechanism of COS. Herein, COSM (MW ≤ 3000 Da) was administered to diet-induced obese mice by oral gavage once daily for eight weeks. The results show that COSM administration reduced body weight; slowed weight gain; reduced serum Glu, insulin, NEFA, TC, TG, and LDL-C levels; increased serum HSL and HDL-C levels; improved inflammation; and reduced lipid droplet size in adipose tissue. Further lipidomic analysis of adipose tissue revealed that 31 lipid species are considered to be underlying lipid biomarkers in COS therapy. These lipids are mainly enriched in pathways involving insulin resistance, thermogenesis, cholesterol metabolism, glyceride metabolism and cyclic adenosine monophosphate (cAMP), which sheds light on the weight loss mechanism of COS. The Western blot assay demonstrated that COSM intervention can improve insulin resistance, inhibit de novo synthesis, and promote thermogenesis and β-oxidation in mitochondria by the AMPK pathway, thereby alleviating high-fat diet-induced obesity. In short, our study can provide a more comprehensive direction for the application of COS in obesity based on molecular markers.
Collapse
Affiliation(s)
- Minchuan Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingqing Huang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Department of Pharmacy, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Jingwen Zhou
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Cuiting Zhi
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd., Science City, Guangzhou 510663, China
| | - Hua Cao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
8
|
Eo H, Kim SH, Ju IG, Huh E, Kim S, Choi JG, Kim SW, Son M, Oh MS. Longan extract suppresses food intake through regulation of POMC/AgRP neuronal activities and endoplasmic reticulum stress in hypothalamus of db/db mice. Front Nutr 2023; 10:1143613. [PMID: 37415911 PMCID: PMC10322219 DOI: 10.3389/fnut.2023.1143613] [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: 01/13/2023] [Accepted: 06/02/2023] [Indexed: 07/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the biggest public health issues worldwide and closely related to development of other chronic diseases such as cardiovascular diseases, cancer and neurodegenerative diseases. Considerable percentage of T2DM patients undergo have suffered from binge eating disorder which exacerbates insulin resistance and metabolic challenges. Longan (Dimocarpus longan L.) and its constituents are reported for their various health benefits. However, it is still unknown whether longan fruit supplementation can ameliorate glucose homeostasis and binge eating disorder found in T2DM. The current study aimed to investigate whether longan fruit extract (LE) supplementation can improve diabetic hyperglycemia through modulation of feeding center located in hypothalamus of db/db T2DM mice. As a result, LE supplementation ameliorated fasting blood glucose levels and reduced excessive epididymal fat accumulation. In addition, LE administration improved glucose tolerance and insulin sensitivity in db/db mice. Especially, LE supplemented mice showed less food consumption which was in line with increase of pro-opiomelanocortin (POMC) neuronal activities and decrease of agouti-related peptide (AgRP) neuronal activities. Furthermore, LE supplementation reduced hypothalamic endoplasmic reticulum (ER) stress which was stimulated in db/db mice. As ER stress is a crucial factor involving in appetite control and glucose homeostasis, the effect of LE supplementation on circulating glucose levels and feeding behavior might be mediated by suppression of hypothalamic ER stress. Collectively, these findings suggest that LE could be a potential nutraceutical for improvement of T2DM as well as patients with satiety issues.
Collapse
Affiliation(s)
- Hyeyoon Eo
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Seong Hye Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - In Gyoung Ju
- Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul, Republic of Korea
| | - Eugene Huh
- Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | - Miwon Son
- MThera Pharma Co., Seoul, Republic of Korea
| | - Myung Sook Oh
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Liu C, Ju R. Manganese-induced neuronal apoptosis: new insights into the role of endoplasmic reticulum stress in regulating autophagy-related proteins. Toxicol Sci 2023; 191:193-200. [PMID: 36519822 DOI: 10.1093/toxsci/kfac130] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Manganese (Mn) is an essential trace element that participates in various physiological and pathological processes. However, epidemiological observations indicate that overexposure to Mn is strongly associated with neurodegenerative disorders and has been recognized as a potential risk factor of neuronal apoptosis. Many mechanisms are involved in the pathogenesis of Mn-induced neuronal apoptosis, such as reactive oxygen species generation, neuroinflammation reactions, protein accumulation, endoplasmic reticulum stress (ER stress), and autophagy, all of which collectively accelerate the process of nerve cell damage. As sophisticated cellular processes for maintaining intracellular homeostasis, ER-mediated unfolded protein response and autophagy both play bilateral roles including cell protection and cell injury under pathophysiological conditions, which might interact with each other. Although emerging evidence suggests that ER stress is involved in regulating the compensatory activation of autophagy to promote cell survival, the inherent relationship between ER stress and autophagy on Mn-induced neurotoxicity remains obscure. Here, our review focuses on discussing the existing mechanisms and connections between ER stress, autophagy, and apoptosis, which provide a new perspective on Mn-induced neuronal apoptosis, and the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu Women's and Children's Central Hospital, Chengdu 611731, China
| | - Rong Ju
- School of Medicine, University of Electronic Science and Technology of China, Chengdu Women's and Children's Central Hospital, Chengdu 611731, China
| |
Collapse
|
10
|
Leptin alleviates endoplasmic reticulum stress induced by cerebral ischemia/reperfusion injury via the PI3K/Akt signaling pathway. Biosci Rep 2022; 42:232083. [PMID: 36367210 PMCID: PMC9744719 DOI: 10.1042/bsr20221443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cerebral ischemic/reperfusion injury (CIRI) is a key factor for the prognosis of ischemic stroke (IS), the leading disease in terms of global disability and fatality rates. Recent studies have shown that endoplasmic reticulum stress (ERS) may be a target against CIRI and that leptin, a peptide hormone, has neuroprotective activity to mitigate CIRI. METHODS An in vitro CIRI model was induced in primary cortical neurons by oxygen-glucose deprivation and reoxygenation (OGD/R) after pretreatment with LY294002 (10 µmol/L) and/or leptin (0.4 mg/L), and cell viability, neuronal morphology and endoplasmic reticulum (ER) dysfunction were evaluated. An in vivo CIRI model was established in rats by middle cerebral artery occlusion and reperfusion (MCAO/R) after the injection of LY294002 (10 μmol/L) and/or leptin (1 mg/kg), and neurological function, infarct volume, cerebral pathological changes, the expression of ERS-related proteins and cell apoptosis were examined. RESULTS In vitro, leptin treatment improved the cell survival rate, ameliorated neuronal pathological morphology and alleviated OGD/R-induced ERS. In vivo, administration of leptin significantly reduced the infarct volume, neurological deficit scores and neuronal apoptosis as well as pathological alterations. In addition, leptin suppressed MCAO/R-induced ERS and may decrease apoptosis by inhibiting ERS-related death and caspase 3 activation. It also regulated expression of the antiapoptotic protein Bcl-2 and the proapoptotic protein Bax in the cortex. Furthermore, the inhibitory effect of leptin on ERS was significantly decreased by the effective phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. CONCLUSIONS These results confirm that ERS plays an important role in CIRI and that leptin can inhibit the activation of ERS through the PI3K/Akt pathway, thereby alleviating CIRI. These findings provide novel therapeutic targets for IS.
Collapse
|
11
|
Demirci Ş, Gün C. Zinc Supplementation Improved Neuropeptide Y, Nesfatin-1, Leptin, C-reactive protein, and HOMA-IR of Diet-Induced Obese Rats. Biol Trace Elem Res 2022; 200:3996-4006. [PMID: 34708332 DOI: 10.1007/s12011-021-02987-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 11/24/2022]
Abstract
Obesity is a mild chronic inflammation that causes many metabolic diseases. It was aimed to investigate some parameters affective on the energy metabolism by adding zinc (Zn, ZnSO4) to drinking water of diet-induced obese rats. Five-week aged, male Sprague Dawley rats divided into as control group, consuming standard rat diet, and high-fat diet (HFD) group. After obesity induced by feeding HFD for 8 weeks, the obese rats were divided into Zn-supplemented obese group (HFD + obese + Zn; 150 mg Zn/L (for 6 weeks), 235 mg Zn/L (7th week), 250 mg Zn/L (8th week) in drinking water) and obese group (HFD + obese). Mean body weight, serum concentrations of C-reactive protein, neuropeptide-Y, leptin, insulin fasting blood glucose, and HOMA-IR were statistically decreased by given Zn in HFD + obese + Zn group compared to HFD + obese rats. It was observed that the total cholesterol, LDL, and HDL cholesterol levels of HFD + obese + Zn group became closer to the control group level, and Zn supplementation caused a statistically significant decrease in cholesterol profile than HFD + obese rats. Also, increased mean serum nesfatin-1 level, an effective protein for the formation of satiety, was analyzed in HFD + obese + Zn group when compared to HFD + obese ones. Serum triglyceride concentration tended to decrease with the effect of Zn in obese rats. In conclusion, it can be said that oral use of Zn could improve energy balance and prevent the occurrence of metabolic diseases related to obesity depending on the anti-inflammatory effect of Zn.
Collapse
Affiliation(s)
- Şule Demirci
- Physiology Department, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Campus, Burdur, Turkey.
| | | |
Collapse
|
12
|
Exercise Improves Spatial Learning and Memory Performance through the Central GLP-1 Receptors. Behav Neurol 2022; 2022:2900628. [PMID: 35774081 PMCID: PMC9239811 DOI: 10.1155/2022/2900628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/05/2022] [Indexed: 11/27/2022] Open
Abstract
The glucagon-like peptide 1 (GLP-1) is a hormone which is produced in the enteroendocrine L-cells in the ileum and the neurons of nucleus tractus solitarius (NTS) in the brain which has numerous metabolic effects. The central GLP-1R's role in cognitive functioning is well known. On the contrary, it has been shown that exercise has positive effects on brain function. So, we decided to elucidate whether the central GLP-1 has a role in memory and learning. Thirty-two rats were used in this experiment in 4 groups. After anesthetizing the rats, the right lateral ventricle was detected, and a cannula was directed to the ventricle. Ten micrograms of exendin-3 or sterile saline, according to the group, was injected via ICV once daily for seven days. The rats in the exercise group considered an exercise period of one hour each day (17 meters per minute) for seven consecutive days. To evaluate the performance of memory and learning, a standard Morris water maze (MWM) tank was utilized. According to the results, the TE-exendin group showed a statistically significant difference from the TE-SAL group in both parameters of latency and time in the zone. In summary, memory and learning were improved by GLP-1R in the exercise group, but not in the sedentary group, which we can hypothesize that exercise can affect memory and learning through this pathway.
Collapse
|
13
|
Kong S, Huang X, Cao H, Bai Y, Che Q, Nie H, Su Z. Anti-obesity effects of galacto-oligosaccharides in obese rats. Eur J Pharmacol 2022; 917:174728. [PMID: 34965390 DOI: 10.1016/j.ejphar.2021.174728] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 12/29/2022]
Abstract
Galacto-oligosaccharides (GOS) are commonly used as prebiotic with a variety of known metabolic benefits; however, whether GOS plays a protective role in obesity remains unknown. Here, we demonstrate that GOS prevented obesity in a rat model of obesity induced by a high-fat diet. Our results showed that GOS effectively slowed weight gain in diet-induced obese rats without affecting energy intake. GOS significantly suppressed the hypertrophy and hyperplasia of white adipose tissue and markedly reduced the ratio of the fat/body. Consistently, GOS significantly improved serum total cholesterol, triglycerides, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol levels, indicating the weight loss activity of GOS. Interestingly, GOS also significantly increased the expression levels of browning proteins, including uncoupling protein 1, peroxisome proliferator-activated receptor-γ, peroxisome proliferator-activated receptor-γ coactivator 1α, and PR domain 16, in both white and brown adipose tissue. Furthermore, we found that GOS markedly increased the expression levels of liver X receptor α, peroxisome proliferation-activated receptor-α, low-density lipoprotein receptor, and cholesterol 7α-hydroxylase proteins in the liver of obese rats. Taken together, we concluded that GOS inhibits obesity by accelerating the browning of white fat cells and the thermogenesis of brown fat cells and that GOS improves host lipid homeostasis by promoting cholesterol catabolism.
Collapse
Affiliation(s)
- Shang Kong
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xingjun Huang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Hua Cao
- Guangdong Cosmetics Engineering & Technology Research Center, School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yan Bai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510310, China
| | - Qishi Che
- Guangzhou Rainhome Pharm & Tech Co., Ltd, Science City, Guangzhou, 510663, China
| | - Hong Nie
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
14
|
Substitution of calorie restriction for protective effects of estrogen on cardiometabolic risk factors and oxidative stress in obese postmenopausal rat model. Life Sci 2022; 294:120367. [DOI: 10.1016/j.lfs.2022.120367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022]
|
15
|
Torres DJ, Pitts MW, Seale LA, Hashimoto AC, An KJ, Hanato AN, Hui KW, Remigio SMA, Carlson BA, Hatfield DL, Berry MJ. Female Mice with Selenocysteine tRNA Deletion in Agrp Neurons Maintain Leptin Sensitivity and Resist Weight Gain While on a High-Fat Diet. Int J Mol Sci 2021; 22:ijms222011010. [PMID: 34681674 PMCID: PMC8539086 DOI: 10.3390/ijms222011010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 11/16/2022] Open
Abstract
The role of the essential trace element selenium in hypothalamic physiology has begun to come to light over recent years. Selenium is used to synthesize a family of proteins participating in redox reactions called selenoproteins, which contain a selenocysteine residue in place of a cysteine. Past studies have shown that disrupted selenoprotein expression in the hypothalamus can adversely impact energy homeostasis. There is also evidence that selenium supports leptin signaling in the hypothalamus by maintaining proper redox balance. In this study, we generated mice with conditional knockout of the selenocysteine tRNA[Ser]Sec gene (Trsp) in an orexigenic cell population called agouti-related peptide (Agrp)-positive neurons. We found that female TrspAgrpKO mice gain less weight while on a high-fat diet, which occurs due to changes in adipose tissue activity. Female TrspAgrpKO mice also retained hypothalamic sensitivity to leptin administration. Male mice were unaffected, however, highlighting the sexually dimorphic influence of selenium on neurobiology and energy homeostasis. These findings provide novel insight into the role of selenoproteins within a small yet heavily influential population of hypothalamic neurons.
Collapse
Affiliation(s)
- Daniel J. Torres
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA; (L.A.S.); (M.J.B.)
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
- Correspondence:
| | - Matthew W. Pitts
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
| | - Lucia A. Seale
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA; (L.A.S.); (M.J.B.)
| | - Ann C. Hashimoto
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
| | - Katlyn J. An
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
| | - Ashley N. Hanato
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
| | - Katherine W. Hui
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
| | - Stella Maris A. Remigio
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA; (M.W.P.); (A.C.H.); (K.J.A.); (A.N.H.); (K.W.H.); (S.M.A.R.)
| | - Bradley A. Carlson
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (B.A.C.); (D.L.H.)
| | - Dolph L. Hatfield
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (B.A.C.); (D.L.H.)
| | - Marla J. Berry
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA; (L.A.S.); (M.J.B.)
| |
Collapse
|
16
|
Liu T, Li T, Chen X, Li Z, Feng M, Yao W, Wan L, Zhang C, Zhang Y. EETs/sEHi alleviates nociception by blocking the crosslink between endoplasmic reticulum stress and neuroinflammation in a central poststroke pain model. J Neuroinflammation 2021; 18:211. [PMID: 34530836 PMCID: PMC8447610 DOI: 10.1186/s12974-021-02255-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 08/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background Central post-stroke pain (CPSP) is a chronic and intolerable neuropathic pain syndrome following a cerebral vascular insult, which negatively impacts the quality of life of stroke survivors but currently lacks efficacious treatments. Though its underlying mechanism remains unclear, clinical features of hyperalgesia and allodynia indicate central sensitization due to excessive neuroinflammation. Recently, the crosslink between neuroinflammation and endoplasmic reticulum (ER) stress has been identified in diverse types of diseases. Nevertheless, whether this interaction contributes to pain development remains unanswered. Epoxyeicosatrienoic acids (EETs)/soluble epoxy hydrolase inhibitors (sEHi) are emerging targets that play a significant role in pain and neuroinflammatory regulation. Moreover, recent studies have revealed that EETs are effective in attenuating ER stress. In this study, we hypothesized that ER stress around the stroke site may activate glial cells and lead to further inflammatory cascades, which constitute a positive feedback loop resulting in central sensitization and CPSP. Additionally, we tested whether EETs/sEHi could attenuate CPSP by suppressing ER stress and neuroinflammation, as well as their vicious cycle, in a rat model of CPSP. Methods Young male SD rats were used to induce CPSP using a model of thalamic hemorrhage and were then treated with TPPU (sEHi) alone or in combination with 14,15-EET or 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE, the EET antagonist), tunicamycin (Tm, ER stress inducer), or 4-PBA (ER stress inhibitor). Nociceptive behaviors, ER stress markers, JNK and p38 (two well-recognized inflammatory kinases of mitogen-activated protein kinase (MAPK) signaling) expression, and glial cell activation were assessed. In addition, some healthy rats were intrathalamically microinjected with Tm or lipopolysaccharide (LPS) to test the interaction between ER stress and neuroinflammation in central pain. Results Analysis of the perithalamic lesion tissue from the brain of CPSP rats demonstrated decreased soluble epoxy hydrolase (sEH) expression, which was accompanied by increased expression of ER stress markers, including BIP, p-IRE, p-PERK, and ATF6. In addition, inflammatory kinases (p-p38 and p-JNK) were upregulated and glial cells were activated. Intrathalamic injection of sEHi (TPPU) increased the paw withdrawal mechanical threshold (PWMT), reduced hallmarks of ER stress and MAPK signaling, and restrained the activation of microglia and astrocytes around the lesion site. However, the analgesic effect of TPPU was completely abolished by 14,15-EEZE. Moreover, microinjection of Tm into the thalamic ventral posterior lateral (VPL) nucleus of healthy rats induced mechanical allodynia and activated MAPK-mediated neuroinflammatory signaling; lipopolysaccharide (LPS) administration led to activation of ER stress along the injected site in healthy rats. Conclusions The present study provides evidence that the interaction between ER stress and neuroinflammation is involved in the mechanism of CPSP. Combined with the previously reported EET/sEHi effects on antinociception and neuroprotection, therapy with agents that target EET signaling may serve as a multi-functional approach in central neuropathic pain by attenuating ER stress, excessive neuroinflammation, and subsequent central sensitization. The use of these agents within a proper time window could not only curtail further nerve injury but also produce an analgesic effect.
Collapse
Affiliation(s)
- Tongtong Liu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Ting Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Xuhui Chen
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Zuofan Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Miaomiao Feng
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Wenlong Yao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Li Wan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Chuanhan Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Yue Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China.
| |
Collapse
|
17
|
Baba B, Caliskan M, Boyuk G, Hacisevki A. Chemical Chaperone PBA Attenuates ER Stress and Upregulates SOCS3 Expression as a Regulator of Leptin Signaling. BIOCHEMISTRY (MOSCOW) 2021; 86:480-488. [PMID: 33941068 DOI: 10.1134/s0006297921040088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Endoplasmic reticulum (ER) is very sensitive to the nutritional and energy states of the cells. Disruption of ER homeostasis leads to the accumulation of unfolded/misfolded proteins in the ER lumen, which is defined as ER stress. ER stress triggers the unfolded protein response (UPR). It is suggested that chronic ER stress is associated with obesity and leptin resistance. We investigated the role of ER stress and the effect of the ER stress inhibitor phenylbutyric acid (PBA) of ER stress, in obesity, as well as their impact on leptin signaling. This study involved twenty-four lean and twenty-four leptin-deficient (ob/ob) mice divided into PBA- and vehicle-treated groups. Pancreatic islets were isolated, incubated with leptin for 48 h, and assayed for the expression of CHOP and XBP1s (UPR signaling indicators) and SOCS3 (regulator of leptin signaling) by RT-qPCR. The expression levels of XBP1s and CHOP were markedly increased in the ob/ob controls compared to other groups with and without leptin treatment. No significant differences in the XBP1s and CHOP expression levels were found between the PBA-treated ob/ob and lean mice. SOCS3 expression was significantly upregulated in the PBA-treated ob/ob mice compared to the ob/ob controls after leptin treatment; but no significant difference in the SOCS3 expression was found between the PBA-treated ob/ob and lean mice with and without leptin treatment. Our findings suggested that ER stress plays an important role in the pathology of obesity, while PBA reduces ER stress and may potentially ameliorate leptin signaling.
Collapse
Affiliation(s)
- Burcu Baba
- Department of Medical Biochemistry, Faculty of Medicine, Yuksek Ihtisas University, Ankara, 06520, Turkey.
| | - Mursel Caliskan
- Department of Genetic, Gaziosmanpaşa Hospital, Yeni Yuzyil University, Istanbul, 34245, Turkey.
| | - Gulbahar Boyuk
- Department of Physiology, Faculty of Medicine, Ankara Medipol University, Ankara, 06050, Turkey.
| | - Aysun Hacisevki
- Department of Biochemistry, Faculty of Pharmacy, Gazi University, Ankara, 06100, Turkey.
| |
Collapse
|
18
|
Impaired Leptin Signalling in Obesity: Is Leptin a New Thermolipokine? Int J Mol Sci 2021; 22:ijms22126445. [PMID: 34208585 PMCID: PMC8235268 DOI: 10.3390/ijms22126445] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Abstract
Leptin is a principal adipose-derived hormone mostly implicated in the regulation of energy balance through the activation of anorexigenic neuronal pathways. Comprehensive studies have established that the maintenance of certain concentrations of circulating leptin is essential to avoid an imbalance in nutrient intake. Indeed, genetic modifications of the leptin/leptin receptor axis and the obesogenic environment may induce changes in leptin levels or action in a manner that accelerates metabolic dysfunctions, resulting in a hyperphagic status and adipose tissue expansion. As a result, a vicious cycle begins wherein hyperleptinaemia and leptin resistance occur, in turn leading to increased food intake and fat enlargement, which is followed by leptin overproduction. In addition, in the context of obesity, a defective thermoregulatory response is associated with impaired leptin signalling overall within the ventromedial nucleus of the hypothalamus. These recent findings highlight the role of leptin in the regulation of adaptive thermogenesis, thus suggesting leptin to be potentially considered as a new thermolipokine. This review provides new insight into the link between obesity, hyperleptinaemia, leptin resistance and leptin deficiency, focusing on the ability to restore leptin sensitiveness by way of enhanced thermogenic responses and highlighting novel anti-obesity therapeutic strategies.
Collapse
|
19
|
The Function of Gastrointestinal Hormones in Obesity-Implications for the Regulation of Energy Intake. Nutrients 2021; 13:nu13061839. [PMID: 34072172 PMCID: PMC8226753 DOI: 10.3390/nu13061839] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of obesity and the challenges of prevention prompted researchers to investigate the mechanisms that control food intake. Food ingestion triggers several physiological responses in the digestive system, including the release of gastrointestinal hormones from enteroendocrine cells that are involved in appetite signalling. Disturbed regulation of gut hormone release may affect energy homeostasis and contribute to obesity. In this review, we summarize the changes that occur in the gut hormone balance during the pre- and postprandial state in obesity and the alterations in the diurnal dynamics of their plasma levels. We further discuss how obesity may affect nutrient sensors on enteroendocrine cells that sense the luminal content and provoke alterations in their secretory profile. Gastric bypass surgery elicits one of the most favorable metabolic outcomes in obese patients. We summarize the effect of different strategies to induce weight loss on gut enteroendocrine function. Although the mechanisms underlying obesity are not fully understood, restoring the gut hormone balance in obesity by targeting nutrient sensors or by combination therapy with gut peptide mimetics represents a novel strategy to ameliorate obesity.
Collapse
|
20
|
Yaribeygi H, Maleki M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Obesity and Insulin Resistance: A Review of Molecular Interactions. Curr Mol Med 2021; 21:182-193. [PMID: 32787760 DOI: 10.2174/1566524020666200812221527] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 07/05/2020] [Accepted: 07/12/2020] [Indexed: 12/06/2022]
Abstract
The prevalence of insulin resistance and diabetes mellitus is rising globally in epidemic proportions. Diabetes and its complications contribute to significant morbidity and mortality. An increase in sedentary lifestyle and consumption of a more energydense diet increased the incidence of obesity which is a significant risk factor for type 2 diabetes. Obesity acts as a potent upstream event that promotes molecular mechanisms involved in insulin resistance and diabetes mellitus. However, the exact molecular mechanisms between obesity and diabetes are not clearly understood. In the current study, we have reviewed the molecular interactions between obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
| | | |
Collapse
|
21
|
Ebbeling CB, Bielak L, Lakin PR, Klein GL, Wong JMW, Luoto PK, Wong WW, Ludwig DS. Energy Requirement Is Higher During Weight-Loss Maintenance in Adults Consuming a Low- Compared with High-Carbohydrate Diet. J Nutr 2020; 150:2009-2015. [PMID: 32470981 PMCID: PMC7398766 DOI: 10.1093/jn/nxaa150] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/18/2019] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Longer-term feeding studies suggest that a low-carbohydrate diet increases energy expenditure, consistent with the carbohydrate-insulin model of obesity. However, the validity of methodology utilized in these studies, involving doubly labeled water (DLW), has been questioned. OBJECTIVE The aim of this study was to determine whether dietary energy requirement for weight-loss maintenance is higher on a low- compared with high-carbohydrate diet. METHODS The study reports secondary outcomes from a feeding study in which the primary outcome was total energy expenditure (TEE). After attaining a mean Run-in weight loss of 10.5%, 164 adults (BMI ≥25 kg/m2; 70.1% women) were randomly assigned to Low-Carbohydrate (percentage of total energy from carbohydrate, fat, protein: 20/60/20), Moderate-Carbohydrate (40/40/20), or High-Carbohydrate (60/20/20) Test diets for 20 wk. Calorie content was adjusted to maintain individual body weight within ± 2 kg of the postweight-loss value. In analyses by intention-to-treat (ITT, completers, n = 148) and per protocol (PP, completers also achieving weight-loss maintenance, n = 110), we compared the estimated energy requirement (EER) from 10 to 20 wk of the Test diets using ANCOVA. RESULTS Mean EER was higher in the Low- versus High-Carbohydrate group in models of varying covariate structure involving ITT [ranging from 181 (95% CI: 8-353) to 246 (64-427) kcal/d; P ≤0.04] and PP [ranging from 245 (43-446) to 323 (122-525) kcal/d; P ≤0.02]. This difference remained significant in sensitivity analyses accounting for change in adiposity and possible nonadherence. CONCLUSIONS Energy requirement was higher on a low- versus high-carbohydrate diet during weight-loss maintenance in adults, commensurate with TEE. These data are consistent with the carbohydrate-insulin model and lend qualified support for the validity of the DLW method with diets varying in macronutrient composition. This trial was registered at clinicaltrials.gov as NCT02068885.
Collapse
Affiliation(s)
- Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston, MA, USA
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Boston, MA, USA
| | - Paul R Lakin
- Institutional Centers for Clinical and Translational Research; Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gloria L Klein
- New Balance Foundation Obesity Prevention Center, Boston, MA, USA
| | - Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Boston, MA, USA
| | - Patricia K Luoto
- Department of Food and Nutrition, Framingham State University, Framingham, MA, USA
| | - William W Wong
- USDA/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine,Houston, TX, USA
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston, MA, USA
| |
Collapse
|
22
|
Deng X, Ye Z, Cao H, Bai Y, Che Q, Guo J, Su Z. Chitosan oligosaccharide ameliorated obesity by reducing endoplasmic reticulum stress in diet-induced obese rats. Food Funct 2020; 11:6285-6296. [PMID: 32602486 DOI: 10.1039/d0fo01107j] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study aimed to determine whether chitosan oligosaccharide (COST) improves overweight by reducing endoplasmic reticulum (ER) stress in the liver and liver cancer cells. METHODS An obesity model was established by feeding Sprague-Dawley rats (ORs) a high-fat diet (HFD) and treating them with COST for 8 weeks. A model of lipid accumulation in hepatocellular carcinoma cells was established by treating HepG2 cells with free fatty acids and COST for 24 h. RESULTS COST treatment of ORs reduced weight gain, inhibited adipose tissue hypertrophy and hyperplasia, and reduced the fat-to-weight ratio. COST improved dyslipidaemia, reduced liver weight and organ index, inhibited hepatic lipid accumulation, and prevented liver steatosis, and the high COST dose increased TC and TG excretion in the stool. Treatment of lipid accumulation in HepG2 cells with COST reduced lipid accumulation and TG levels. COST modulated the expression of genes related to fat metabolism and ER stress response pathway-related factors in liver tissue and HepG2 cells. CONCLUSIONS COST can inhibit weight gain and improve dyslipidaemia and lipid metabolism in ORs. The COST-mediated regulation of hepatic and HepG2 cell lipid metabolism might be related to inhibition of fat synthesis, acceleration of lipid oxidative catabolism and reduction in ER stress.
Collapse
Affiliation(s)
- Xiaoyi Deng
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou (510006), China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Tan Q, Liu Y, Deng X, Chen J, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Autophagy: a promising process for the treatment of acetaminophen-induced liver injury. Arch Toxicol 2020; 94:2925-2938. [PMID: 32529281 DOI: 10.1007/s00204-020-02780-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
Toxicity from drugs has become an important cause of acute liver failure. Acetaminophen, a commonly used analgesic, can cause severe acute liver injury that can worsen into acute liver failure. Autophagy, a protective cell programme, has been reported to have protective effects in a variety of diseases such as cancer, immune diseases, neurodegenerative diseases, and inflammatory diseases. In this review, we describe how an excess of acetaminophen causes liver injury step by step, from the formation of the initial protein adduct to the final hepatocyte necrosis, as well as the induction of autophagy and its beneficial effects on diseases. Emphasis is placed on the potential effect of autophagy on improving the damage of acetaminophen to hepatocytes. Finally, we are committed to providing insights into the treatment of acute liver failure through the mechanism of acetaminophen induced liver injury, the mechanism of autophagy, and the link between autophagy and liver injury.
Collapse
Affiliation(s)
- Qiuhua Tan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongjian Liu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoyi Deng
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiajia Chen
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) CO., Ltd., New Taipei City, Taiwan, ROC
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) CO., Ltd., New Taipei City, Taiwan, ROC
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology CO. Ltd., Yunfu, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
24
|
Li X, Shi W, Xiong Q, Hu Y, Qin X, Wan G, Zeng Q. Leptin improves intestinal flora dysfunction in mice with high-fat diet-induced obesity. J Int Med Res 2020; 48:300060520920062. [PMID: 32529880 PMCID: PMC7294385 DOI: 10.1177/0300060520920062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE This study investigated the effects of leptin on intestinal flora and inflammation in mice with high-fat diet (HFD)-induced obesity. METHODS Mice were fed an HFD for 8 weeks; some were concurrently administered oral leptin for 4 weeks. Pathological changes in adipose tissue were detected using hematoxylin-eosin staining; endotoxin content in adipose tissue was measured by enzyme-linked immunosorbent assay. Intestinal flora were characterized by 16S bacterial rDNA sequencing. Levels of Toll-like receptor 4 (TLR4), nuclear factor-κB inhibitor α (IκB-α), and phosphorylated c-Jun N-terminal kinase (p-JNK) were detected by western blotting. RESULTS Mice in the HFD group exhibited weight gain, elevated endotoxin content, and adipocyte hypertrophy, compared with the non-obese control group. Moreover, abundance of bacteria in the Bacteroides genus and community diversity were both reduced in the HFD group; reductions also were observed at corresponding phylum, class, and order levels. Levels of TLR4, IκB-α, and p-JNK were also elevated in the HFD group. Compared with the model group, leptin administration reduced the weight gain and endotoxin content, while increasing Bacteroides abundance and community diversity; it also reduced the levels of TLR4, IκB-α, and p-JNK. CONCLUSION Leptin administration improved intestinal flora dysfunction and inflammation in mice with HFD-induced obesity.
Collapse
Affiliation(s)
- Xiaolin Li
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| | - Weihong Shi
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| | - Qinghua Xiong
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| | - Yungang Hu
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| | - Xu Qin
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| | - Guanqun Wan
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| | - Qi Zeng
- Department of Plastic Maxillofacial Surgery, People’s Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
25
|
Wang J, He W, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Mutual interaction between endoplasmic reticulum and mitochondria in nonalcoholic fatty liver disease. Lipids Health Dis 2020; 19:72. [PMID: 32284046 PMCID: PMC7155254 DOI: 10.1186/s12944-020-01210-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common metabolic syndrome. Imbalances between liver lipid output and input are the direct causes of NAFLD, and hepatic steatosis is the pathological premise and basis for NAFLD progression. Mutual interaction between endoplasmic reticulum stress (ERS) and oxidative stress play important roles in NAFLD pathogenesis. Notably, mitochondria-associated membranes (MAMs) act as a structural bridges for functional clustering of molecules, particularly for Ca2+, lipids, and reactive oxygen species (ROS) exchange. Previous studies have examined the crucial roles of ERS and ROS in NAFLD and have shown that MAM structural and functional integrity determines normal ER- mitochondria communication. Upon disruption of MAM integrity, miscommunication directly or indirectly causes imbalances in Ca2+ homeostasis and increases ERS and oxidative stress. Here, we emphasize the involvement of MAMs in glucose and lipid metabolism, chronic inflammation and insulin resistance in NAFLD and summarize MAM-targeting drugs and compounds, most of which achieve their therapeutic or ameliorative effects on NAFLD by improving MAM integrity. Therefore, targeting MAMs may be a viable strategy for NAFLD treatment. This review provides new ideas and key points for basic NAFLD research and drug development centred on mitochondria and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Jin Wang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wanping He
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) Co., LTD, 2F.-1, No. 250, Zhongshan Rd., Linkou Dist, New Taipei City, 24446, Taiwan
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology Co, Ltd Swan-kan-chiau Ind. Dist., Kaofong Village, Yunfu City, Guangdong, China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Key Laboratory of Modulating Liver to Treat Hyperlipemia SATCM, Level 3 Laboratory of Lipid Metabolism SATCM, Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
26
|
Yao Y. Ginsenosides reduce body weight and ameliorate hepatic steatosis in high fat diet‑induced obese mice via endoplasmic reticulum stress and p‑STAT3/STAT3 signaling. Mol Med Rep 2020; 21:1059-1070. [PMID: 32016448 PMCID: PMC7003045 DOI: 10.3892/mmr.2020.10935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 10/24/2019] [Indexed: 12/28/2022] Open
Abstract
Obesity has been increasing globally for over three decades. According to previous studies, dietary obesity is usually associated with endoplasmic reticulum stress (ERS) and STAT3 signaling, which result in interference with the homeostatic control of energy and lipid metabolism. Ginsenosides (GS) administered to mice will modulate adiposity and food intake; however, the mechanism of food inhibition is unknown. The aim of the present study was to investigate whether GS may inhibit ERS and regulate STAT3 phosphorylation in GT1‑7 cells (a mouse hypothalamus gonadotropin‑releasing hormone neuron cell line) and the hypothalamus in order to reduce the body weight and ameliorate hepatic steatosis in high fat diet (HFD)‑induced obese mice. In the present study, GS inhibited the appetite, reduced the body weight, visceral fat, body fat content and blood glucose, and ameliorated the glucose tolerance of the obese mice compared with HFD mice. In addition, the levels of aspartate aminotransferase and alanine aminotransferase, triglyceride (TG), leptin and insulin in the serum were reduced compared with HFD mice. There was less TG in the liver, but more in the feces compared with HFD mice. Using hematoxylin and eosin staining of HepG2 cells and liver tissues, GS were demonstrated to improve the non‑alcoholic fatty liver of the HFD‑induced obese mice and reduce the diameter of the fat cells compared with HFD mice. GS also increased oxygen consumption and carbon dioxide emissions in the metabolic cage data compared with HFD mice. In the GT1‑7 cells, GS alleviated the ERS induced by tunicamycin and enhanced the activation of the STAT3 phosphorylation pathway. Furthermore the ERS of the liver was relieved to achieve the aforementioned pharmacological effects. GS were used in the homeostatic control of the energy and lipid metabolism of a diet‑induced obesity model. In conclusion, present studies suggest that GS exert these effects by increasing STAT3 phosphorylation expression and reducing the ERS. Thus, GS reduce body weight and ameliorate hepatic steatosis in HFD‑induced obese mice.
Collapse
Affiliation(s)
- Yin Yao
- Department of Traditional Chinese Medicine Chemistry, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 200000, P.R. China
| |
Collapse
|
27
|
Liu Y, Chen J, Tan Q, Deng X, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Nondigestible Oligosaccharides with Anti-Obesity Effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:4-16. [PMID: 31829005 DOI: 10.1021/acs.jafc.9b06079] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Obesity has an important influence on health conditions, causing a multitude of complications and comorbidities, and drug therapy is considered to be one of the treatment strategies. Nowadays, there is increasing interest in the study of intestinal microbiota regulation of obesity; also, an increasing number of agricultural and sideline products have been found to have anti-obesity potential. In the present review, we summarize an overview of current known and potential anti-obesity oligosaccharides and their molecular structures. We describe their anti-obesity potential activity and the molecular structure associated with this activity, the regulation of intestinal microbiota composition and its mechanism of action, including regulation of the short-chain fatty acid (SCFA) pathway and altering bile acid (BA) pathway. This review will provide new ideas for us to develop new anti-obesity functional foods.
Collapse
Affiliation(s)
- Yongjian Liu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Jiajia Chen
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Qiuhua Tan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Xiaoyi Deng
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) CO., LTD. , Linkou District, New Taipei City 24446 , Taiwan China
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) CO., LTD. , Linkou District, New Taipei City 24446 , Taiwan China
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology CO., LTD. , Swan-kan-chiau Industrial District, Kaofong Village Yunfu City 527343 , Guangdong , China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| |
Collapse
|
28
|
Liu Y, Yu J, Shi YC, Zhang Y, Lin S. The role of inflammation and endoplasmic reticulum stress in obesity-related cognitive impairment. Life Sci 2019; 233:116707. [PMID: 31374234 DOI: 10.1016/j.lfs.2019.116707] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/16/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
The epidemiological investigations and animal model experiments have confirmed the impact of obesity on the brain, behavior, and cognition. However, the mechanism by which obesity affects cognitive function is not fully understood. With the development of an aging society, there is an increase in the economic and social burden caused by the decline in cognitive function. This manuscript reviews the effects of inflammation and endoplasmic reticulum stress (ERS) on the hypothalamus, hippocampus, and the possible impact on cognitive impairment. These findings provide new insights into the pathophysiological mechanisms that lead to the development of cognitive impairment in the context of obesity.
Collapse
Affiliation(s)
- Yilan Liu
- Quanzhou First Hospital, Fujian Medical University, China
| | - Jing Yu
- Quanzhou First Hospital, Fujian Medical University, China
| | - Yan-Chuan Shi
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), China
| | - Yi Zhang
- Quanzhou First Hospital, Fujian Medical University, China.
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), China; Illawarra Health and Medical Research Institute, Wollongong 2522, Australia.
| |
Collapse
|
29
|
Effect of Konjac Mannan Oligosaccharides on Glucose Homeostasis via the Improvement of Insulin and Leptin Resistance In Vitro and In Vivo. Nutrients 2019; 11:nu11081705. [PMID: 31344867 PMCID: PMC6723648 DOI: 10.3390/nu11081705] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
Functional oligosaccharides, particularly konjac mannan oligosaccharides (KMOS), can regulate glucose metabolism. However, the molecular mechanisms involved in the hypoglycemic effect of KMOS remain largely unknown. Here, the effect of KMOS supplementation on glucose homeostasis was evaluated in both high-fat diet (HFD)-fed C57BL/6J mice and high-glucosamine-induced HepG2 cells. KMOS supplementation remarkably ameliorated the fasting blood glucose, glucose tolerance, and insulin tolerance of HFD-fed mice. Abnormalities of triglyceride and glycogen metabolism in the liver induced by the HFD were reversed by KMOS supplementation. The insulin signaling pathway was activated by KMOS, with stimulation of GLUT2 membrane translocation and glucose uptake in HepG2 cells via the AMPK pathway. Moreover, KMOS suppressed p-mTOR expression and stimulated the GSK-3β/CREB pathway via the AMPK pathway. KMOS significantly upregulated leptin receptor expression and downregulated PTP1B and SOCS3 levels in the liver and brain, with a decreased serum leptin concentration. Phosphorylation of JAK2 and STAT3 in the liver was activated by KMOS supplementation, while the expressions of Sirt1, Tfam, and Pgc1-α in the brain were elevated. Conclusively, KMOS attenuated HFD-induced glucose metabolism dysfunction through the regulation of insulin resistance and leptin resistance. This finding indicates that KMOS have potential value as an anti-hyperglycemic dietary supplement.
Collapse
|
30
|
The Role of Protein Tyrosine Phosphatase (PTP)-1B in Cardiovascular Disease and Its Interplay with Insulin Resistance. Biomolecules 2019; 9:biom9070286. [PMID: 31319588 PMCID: PMC6680919 DOI: 10.3390/biom9070286] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/06/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022] Open
Abstract
Endothelial dysfunction is a key feature of cardiovascular disorders associated with obesity and diabetes. Several studies identified protein tyrosine phosphatase (PTP)-1B, a member of the PTP superfamily, as a major negative regulator for insulin receptor signaling and a novel molecular player in endothelial dysfunction and cardiovascular disease. Unlike other anti-diabetic approaches, genetic deletion or pharmacological inhibition of PTP1B was found to improve glucose homeostasis and insulin signaling without causing lipid buildup in the liver, which represents an advantage over existing therapies. Furthermore, PTP1B was reported to contribute to cardiovascular disturbances, at various molecular levels, which places this enzyme as a unique single therapeutic target for both diabetes and cardiovascular disorders. Synthesizing selective small molecule inhibitors for PTP1B is faced with multiple challenges linked to its similarity of sequence with other PTPs; however, overcoming these challenges would pave the way for novel approaches to treat diabetes and its concurrent cardiovascular complications. In this review article, we summarized the major roles of PTP1B in cardiovascular disease with special emphasis on endothelial dysfunction and its interplay with insulin resistance. Furthermore, we discussed some of the major challenges hindering the synthesis of selective inhibitors for PTP1B.
Collapse
|
31
|
Salonia A, Rastrelli G, Hackett G, Seminara SB, Huhtaniemi IT, Rey RA, Hellstrom WJG, Palmert MR, Corona G, Dohle GR, Khera M, Chan YM, Maggi M. Paediatric and adult-onset male hypogonadism. Nat Rev Dis Primers 2019; 5:38. [PMID: 31147553 PMCID: PMC6944317 DOI: 10.1038/s41572-019-0087-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The hypothalamic-pituitary-gonadal axis is of relevance in many processes related to the development, maturation and ageing of the male. Through this axis, a cascade of coordinated activities is carried out leading to sustained testicular endocrine function, with gonadal testosterone production, as well as exocrine function, with spermatogenesis. Conditions impairing the hypothalamic-pituitary-gonadal axis during paediatric or pubertal life may result in delayed puberty. Late-onset hypogonadism is a clinical condition in the ageing male combining low concentrations of circulating testosterone and specific symptoms associated with impaired hormone production. Testosterone therapy for congenital forms of hypogonadism must be lifelong, whereas testosterone treatment of late-onset hypogonadism remains a matter of debate because of unclear indications for replacement, uncertain efficacy and potential risks. This Primer focuses on a reappraisal of the physiological role of testosterone, with emphasis on the critical interpretation of the hypogonadal conditions throughout the lifespan of the male individual, with the exception of hypogonadal states resulting from congenital disorders of sex development.
Collapse
Affiliation(s)
- Andrea Salonia
- Division of Experimental Oncology, Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Giulia Rastrelli
- Sexual Medicine and Andrology Unit Department of Experimental Clinical and Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Geoffrey Hackett
- Department of Urology, University of Bedfordshire, Bedfordshire, UK
| | - Stephanie B Seminara
- Harvard Reproductive Sciences Center and Reproductive Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ilpo T Huhtaniemi
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, UK
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños R. Gutiérrez, Buenos Aires, Argentina
| | - Wayne J G Hellstrom
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mark R Palmert
- Division of Endocrinology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Giovanni Corona
- Sexual Medicine and Andrology Unit Department of Experimental Clinical and Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
- Endocrinology Unit, Medical Department, Azienda Usl Bologna Maggiore-Bellaria Hospital, Bologna, Italy
| | - Gert R Dohle
- Department of Urology, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Mohit Khera
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - Yee-Ming Chan
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Mario Maggi
- Sexual Medicine and Andrology Unit Department of Experimental Clinical and Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
- Istituto Nazionale Biostrutture e Biosistemi (INBB), Rome, Italy
| |
Collapse
|
32
|
Wang X, Chen D, Li Y, Zhao S, Chen C, Ning D. Alleviating effects of walnut green husk extract on disorders of lipid levels and gut bacteria flora in high fat diet-induced obesity rats. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
33
|
Ebbeling CB, Feldman HA, Klein GL, Wong JMW, Bielak L, Steltz SK, Luoto PK, Wolfe RR, Wong WW, Ludwig DS. Effects of a low carbohydrate diet on energy expenditure during weight loss maintenance: randomized trial. BMJ 2018; 363:k4583. [PMID: 30429127 PMCID: PMC6233655 DOI: 10.1136/bmj.k4583] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To determine the effects of diets varying in carbohydrate to fat ratio on total energy expenditure. DESIGN Randomized trial. SETTING Multicenter collaboration at US two sites, August 2014 to May 2017. PARTICIPANTS 164 adults aged 18-65 years with a body mass index of 25 or more. INTERVENTIONS After 12% (within 2%) weight loss on a run-in diet, participants were randomly assigned to one of three test diets according to carbohydrate content (high, 60%, n=54; moderate, 40%, n=53; or low, 20%, n=57) for 20 weeks. Test diets were controlled for protein and were energy adjusted to maintain weight loss within 2 kg. To test for effect modification predicted by the carbohydrate-insulin model, the sample was divided into thirds of pre-weight loss insulin secretion (insulin concentration 30 minutes after oral glucose). MAIN OUTCOME MEASURES The primary outcome was total energy expenditure, measured with doubly labeled water, by intention-to-treat analysis. Per protocol analysis included participants who maintained target weight loss, potentially providing a more precise effect estimate. Secondary outcomes were resting energy expenditure, measures of physical activity, and levels of the metabolic hormones leptin and ghrelin. RESULTS Total energy expenditure differed by diet in the intention-to-treat analysis (n=162, P=0.002), with a linear trend of 52 kcal/d (95% confidence interval 23 to 82) for every 10% decrease in the contribution of carbohydrate to total energy intake (1 kcal=4.18 kJ=0.00418 MJ). Change in total energy expenditure was 91 kcal/d (95% confidence interval -29 to 210) greater in participants assigned to the moderate carbohydrate diet and 209 kcal/d (91 to 326) greater in those assigned to the low carbohydrate diet compared with the high carbohydrate diet. In the per protocol analysis (n=120, P<0.001), the respective differences were 131 kcal/d (-6 to 267) and 278 kcal/d (144 to 411). Among participants in the highest third of pre-weight loss insulin secretion, the difference between the low and high carbohydrate diet was 308 kcal/d in the intention-to-treat analysis and 478 kcal/d in the per protocol analysis (P<0.004). Ghrelin was significantly lower in participants assigned to the low carbohydrate diet compared with those assigned to the high carbohydrate diet (both analyses). Leptin was also significantly lower in participants assigned to the low carbohydrate diet (per protocol). CONCLUSIONS Consistent with the carbohydrate-insulin model, lowering dietary carbohydrate increased energy expenditure during weight loss maintenance. This metabolic effect may improve the success of obesity treatment, especially among those with high insulin secretion. TRIAL REGISTRATION ClinicalTrials.gov NCT02068885.
Collapse
Affiliation(s)
- Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Henry A Feldman
- Harvard Medical School, Boston, MA, USA
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | - Gloria L Klein
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Sarah K Steltz
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Patricia K Luoto
- Department of Food and Nutrition, Framingham State University, Framingham, MA, USA
| | - Robert R Wolfe
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - William W Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
Potential Involvement of Peripheral Leptin/STAT3 Signaling in the Effects of Resveratrol and Its Metabolites on Reducing Body Fat Accumulation. Nutrients 2018; 10:nu10111757. [PMID: 30441779 PMCID: PMC6265754 DOI: 10.3390/nu10111757] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
Bioactive compounds such as polyphenols have increased in importance in recent years, and among them, resveratrol (3,5,4′-trihydroxy-trans-stilbene) has generated great interest as an anti-obesity agent. Recent investigations have highlighted the importance of leptin signaling in lipid metabolism in peripheral organs. The aims of this study were (1) to investigate whether resveratrol can reduce fat accumulation in peripheral tissues by increasing their leptin sensitivity and (2) to identify which resveratrol-derived circulating metabolites are potentially involved in these metabolic effects. Serum leptin levels and the leptin signaling pathway were assessed in diet-induced obese rats. Moreover, serum metabolites of resveratrol were studied by ultra-high performance liquid chromatography–mass spectrometry (UHPLC-MSn). The daily consumption of 200 mg/kg of resveratrol, but not doses of 50 and 100 mg/kg, reduced body weight and fat accumulation in obese rats and restored leptin sensitivity in the periphery. These effects were due to increases in sirtuin 1 activity in the liver, leptin receptors in muscle and protection against endoplasmic reticulum (ER)-stress in adipose tissue. In general, the resveratrol metabolites associated with these beneficial effects were derived from both phase II and microbiota metabolism, although only those derived from microbiota increased proportionally with the administered dose of resveratrol. In conclusion, resveratrol reversed leptin resistance caused by diet-induced obesity in peripheral organs using tissue-specific mechanisms.
Collapse
|
35
|
Ceylan AF, Wang S, Kandadi MR, Chen J, Hua Y, Pei Z, Nair S, Ren J. Cardiomyocyte-specific knockout of endothelin receptor a attenuates obesity cardiomyopathy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3339-3352. [DOI: 10.1016/j.bbadis.2018.07.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/14/2018] [Accepted: 07/16/2018] [Indexed: 12/20/2022]
|
36
|
Endoplasmic Reticulum Stress in Metabolic Disorders. Cells 2018; 7:cells7060063. [PMID: 29921793 PMCID: PMC6025008 DOI: 10.3390/cells7060063] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Metabolic disorders have become among the most serious threats to human health, leading to severe chronic diseases such as obesity, type 2 diabetes, and non-alcoholic fatty liver disease, as well as cardiovascular diseases. Interestingly, despite the fact that each of these diseases has different physiological and clinical symptoms, they appear to share certain pathological traits such as intracellular stress and inflammation induced by metabolic disturbance stemmed from over nutrition frequently aggravated by a modern, sedentary life style. These modern ways of living inundate cells and organs with saturating levels of sugar and fat, leading to glycotoxicity and lipotoxicity that induce intracellular stress signaling ranging from oxidative to ER stress response to cope with the metabolic insults (Mukherjee, et al., 2015). In this review, we discuss the roles played by cellular stress and its responses in shaping metabolic disorders. We have summarized here current mechanistic insights explaining the pathogenesis of these disorders. These are followed by a discussion of the latest therapies targeting the stress response pathways.
Collapse
|
37
|
Pan H, Fu C, Huang L, Jiang Y, Deng X, Guo J, Su Z. Anti-Obesity Effect of Chitosan Oligosaccharide Capsules (COSCs) in Obese Rats by Ameliorating Leptin Resistance and Adipogenesis. Mar Drugs 2018; 16:md16060198. [PMID: 29874843 PMCID: PMC6025060 DOI: 10.3390/md16060198] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/20/2018] [Accepted: 06/01/2018] [Indexed: 12/15/2022] Open
Abstract
Obesity is a global disease that causes many metabolic disorders. However, effective agents for the prevention or treatment of obesity remain limited. This study investigated the anti-obesity effect and mechanism of chitosan oligosaccharide capsules (COSCs) on rats suffering from obesity induced by a high-fat diet (HFD). After the eight-week administration of COSCs on obese rats, the body weight gain, fat/body ratio, and related biochemical indices were measured. The hepatic expressions of the leptin signal pathway (JAK2-STAT3) and gene expressions of adipogenesis-related targets were also determined. Our data showed that COSCs can regulate body weight gain, lipids, serum alanine aminotransferase, and aspartate aminotransferase, as well as upregulate the hepatic leptin receptor-b (LepRb) and the phosphorylation of JAK2 and STAT3. Meanwhile, marked increased expressions of liver sterol regulatory element-binding protein-1c, fatty acid synthase, acetyl-CoA carboxylase, 3-hydroxy-3-methylglutaryl-CoA reductase, adiponectin, adipose peroxisome proliferator-activated receptor γ, CCAAT-enhancer binding protein α, adipose differentiation-related protein, and SREBP-1c were observed. The results suggested that COSCs activate the JAK2-STAT3 signaling pathway to alleviate leptin resistance and suppress adipogenesis to reduce lipid accumulation. Thus, they can potentially be used for obesity treatment.
Collapse
Affiliation(s)
- Haitao Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China.
| | - Chuhan Fu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lanlan Huang
- Guangdong Food and Drug Vocational Technical School, Guangzhou 510663, China.
| | - Yao Jiang
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Xiaoyi Deng
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs, Guangdong Pharmaceutical University, Guangzhou 510006, China.
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|