1
|
Ma P, Ou H, Cai J, Zhang Y, Ou Y. DRD2-Mediated AMPK Ubiquitination Regulates the Occurrence of Hepatic Steatosis. Liver Int 2025; 45:e70053. [PMID: 40052721 DOI: 10.1111/liv.70053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND & AIMS G protein-coupled receptors (GPCRs) are important potential drug targets for the treatment of metabolic disorders. The D2 dopamine receptor (DRD2), a GPCR receptor, is a member of the dopamine receptor family. However, the role of DRD2 in regulating lipid metabolism, especially in hepatic steatosis, is unclear. METHODS Eight-week male mice were fed HFHC/MCD to induce the MASH model. AAV2/8 containing the TBG promoter was used to knock down and overexpress DRD2 in mouse liver. Co-immunoprecipitation, Western lotting, immunofluorescence, and immunohistochemistry were used to investigate the mechanisms and screen DRD2 antagonists. RESULTS The study found that activation of PKC leads to the elevation and internalisation of DRD2 in a high-fat environment. Knockdown of DRD2 in mouse liver can effectively interfere with the progression of MASH, while overexpression of DRD2 significantly aggravates the process of MASH. The study on the mechanism of DRD2 regulating lipid metabolism found that the internalisation of DRD2 could lead to dephosphorylation of pAKT (T308) by binding to β-arrestin2 and pAKT, thereby inducing ubiquitin-dependent degradation of AMPK and exacerbating steatosis. L-741626, a DRD2 antagonist, was found to interfere with the internalisation of DRD2 in a high-fat environment. It has been shown that L-741626 can treat MASH by regulating the AKT-AMPK signalling axis in vitro and in vivo. CONCLUSIONS In conclusion, this study demonstrated that internalisation of DRD2 in a high-fat environment aggravated MASH progression through the AKT-AMPK signalling axis. Furthermore, L-741626, as a DRD2 antagonist, has the potential to treat MASH.
Collapse
Affiliation(s)
- Peng Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hao Ou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junze Cai
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yuanli Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yu Ou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Liu S, Li F, Cai Y, Sun L, Ren L, Yin M, Cui H, Pan Y, Gang X, Wang G. Gout drives metabolic dysfunction-associated steatotic liver disease through gut microbiota and inflammatory mediators. Sci Rep 2025; 15:9395. [PMID: 40102566 PMCID: PMC11920238 DOI: 10.1038/s41598-025-94118-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
This study explores the relationship between gout and metabolic dysfunction-associated steatotic liver disease (MASLD), two metabolic conditions linked to worsening health outcomes. While hyperuricemia's association with MASLD is established, the specific connection between gout and MASLD remains less explored. Using data from the UK Biobank, the study employs COX proportional hazard models, multi-state survival analysis, and Mendelian randomization to assess the independent and mutual risks of gout and MASLD. Findings indicate a mutual risk increase: male gout patients, those younger than 60, and those with high BMI are particularly susceptible to MASLD, while female MASLD patients are at heightened risk for gout. Shared risk factors for both conditions include high BMI, hypertension, diabetes, and hyperuricemia. The study further identifies a bidirectional causal link, with gout leading to MASLD, mediated by gut microbiota Ruminococcaceae and proteins like IL-2 and GDF11, implicating specific metabolic pathways. The findings highlight a clinical and mechanistic correlation, emphasizing the need for targeted interventions to address these overlapping metabolic pathways in future treatments.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Fan Li
- Department of Hepatobiliary and Pancreatic Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yunjia Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Linan Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Mengsha Yin
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Huijuan Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yujie Pan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
3
|
Kubota N, Kubota T, Kadowaki T. Physiological and pathophysiological actions of insulin in the liver. Endocr J 2025; 72:149-159. [PMID: 39231651 PMCID: PMC11850106 DOI: 10.1507/endocrj.ej24-0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/21/2024] [Indexed: 09/06/2024] Open
Abstract
The liver plays an important role in the control of glucose homeostasis. When insulin levels are low, such as in the fasting state, gluconeogenesis and glycogenolysis are stimulated to maintain the blood glucose levels. Conversely, in the presence of increased insulin levels, such as after a meal, synthesis of glycogen and lipid occurs to maintain the blood glucose levels within normal range. Insulin receptor signaling regulates glycogenesis, gluconeogenesis and lipogenesis through downstream pathways such as the insulin receptor substrate (IRS)-phosphoinositide 3 (PI3) kinase-Akt pathway. IRS-1 and IRS-2 are abundantly expressed in the liver and are thought to be responsible for transmitting the insulin signal from the insulin receptor to the intracellular effectors involved in the regulation of glucose and lipid homeostasis. Impaired insulin receptor signaling can cause hepatic insulin resistance and lead to type 2 diabetes. In the present study, we focus on a concept called "selective insulin resistance," which has received increasing attention recently: the frequent coexistence of hyperglycemia and hepatic steatosis in people with type 2 diabetes and obesity suggests that it is possible for the insulin signaling regulating gluconeogenesis to be impaired even while that regulating lipogenesis is preserved, suggestive of selective insulin resistance. In this review, we review the progress in research on the insulin actions and insulin signaling in the liver.
Collapse
Affiliation(s)
- Naoto Kubota
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tetsuya Kubota
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo 103-0002, Japan
| | | |
Collapse
|
4
|
Tambe V, Soderblom EJ, Kayesh R, Aditya V, Xu C, Yue W. Regulation of organic anion transporting polypeptide 1B1 transport function by concurrent phosphorylation and lysine-acetylation: A novel posttranslational regulation mechanism. Mol Pharmacol 2025; 107:100007. [PMID: 40023514 PMCID: PMC11934288 DOI: 10.1016/j.molpha.2024.100007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B1 is crucial for hepatic uptake of many drugs and endogenous substrates. The clinically relevant OATP1B1 c.521 T>C (V174A) polymorphism exhibits reduced transport activity in vitro and in vivo in humans. Previously, we reported increased total phosphorylation of V174A-OATP1B1 compared to wild-type (WT)-OATP1B1, although the differentially phosphorylated sites remain to be identified. Lysine-acetylation, a key posttranslational modification (PTM), has not been investigated in OATP1B1. This study aimed to identify differential PTMs of WT-OATP1B1 and V174A-OATP1B1 by quantitatively comparing the relative abundance of modified peptides using liquid chromatography-tandem mass spectrometry-based proteomics and to assess the impact of these PTMs on OATP1B1 transport function using [3H]-estradiol-17-β-D-glucuronide as substrate in transporter-expressing human embryonic kidney 293 cells. We discovered that OATP1B1 is lysine-acetylated at 13 residues. Compared to WT-OATP1B1, V174A-OATP1B1 has increased concurrent phosphorylation at S659 and S663 and concurrent phosphorylation (at S659 and S663) and lysine-acetylation (at K650) (P < .05). Variants mimicking concurrent phosphorylation (S659E-S663E-OATP1B1) and concurrent phosphorylation and acetylation (K650Q-659E-S663E-OATP1B1) both demonstrated reduced substrate transport by 0.86 ± 0.055-fold and 0.65 ± 0.047-fold of WT-OATP1B1 (both P < .05), respectively. Single-site mimics of phosphorylation or lysine-acetylation at K650, S659, and S663 did not affect OATP1B1 transport function, indicating cooperative effects on OATP1B1 by concurrent PTMs. All variants and WT-OATP1B1 were primarily localized to the plasma membrane and colocalized with plasma membrane protein Na/K-ATPase as determined by immunofluorescent staining and confocal microscopy. The current study elucidates a novel mechanism in which concurrent serine-phosphorylation and lysine-acetylation impair OATP1B1-mediated transport, suggesting potential interplay between these PTMs in regulating OATP1B1. SIGNIFICANCE STATEMENT: Understanding organic anion transporting polypeptide (OATP1B1) regulation is key to predicting altered drug disposition. The Val174Ala-OATP1B1 polymorphism exhibits reduced transport activity and is the most effective predictor of statin-induced myopathy. Val174Ala-OATP1B1 was found to be associated with increased serine-phosphorylation at Ser659 and Ser663 and lysine-acetylation at Lys650; concurrent PTMs at these sites reduce OATP1B1 function. These findings revealed a novel mechanism involved in transporter regulation, suggesting potential interplay between these PTMs in governing hepatic drug transport and response.
Collapse
Affiliation(s)
- Vishakha Tambe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Erik J Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, North Carolina
| | - Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Vikram Aditya
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences, Oklahoma City, Oklahoma.
| |
Collapse
|
5
|
Ketema EB, Lopaschuk GD. The Impact of Obesity on Cardiac Energy Metabolism and Efficiency in Heart Failure With Preserved Ejection Fraction. Can J Cardiol 2025:S0828-282X(25)00099-6. [PMID: 39892611 DOI: 10.1016/j.cjca.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence and prevalence of heart failure with preserved ejection fraction (HFpEF) continues to rise, and now comprises more than half of all heart failure cases. There are many risk factors for HFpEF, including older age, hypertension, diabetes, dyslipidemia, sedentary behaviour, and obesity. The rising prevalence of obesity in society is a particularly important contributor to HFpEF development and severity. Obesity can adversely affect the heart, including inducing marked alterations in cardiac energy metabolism. This includes obesity-induced impairments in mitochondrial function, and an increase in fatty acid uptake and mitochondrial fatty acid β-oxidation. This increase in myocardial fatty acid metabolism is accompanied by an impaired myocardial insulin signaling and a marked decrease in glucose oxidation. This switch from glucose to fatty acid metabolism decreases cardiac efficiency and can contribute to severity of HFpEF. Increased myocardial fatty acid uptake in obesity is also associated with the accumulation of fatty acids, resulting in cardiac lipotoxicity. Obesity also results in dramatic changes in the release of adipokines, which can negatively impact cardiac function and energy metabolism. Obesity-induced increases in epicardial fat can also increase cardiac insulin resistance and negatively affect cardiac energy metabolism and HFpEF. However, optimizing cardiac energy metabolism in obese subjects may be one approach to preventing and treating HFpEF. This review discusses what is presently known about the effects of obesity on cardiac energy metabolism and insulin signaling in HFpEF. The clinical implications of obesity and energy metabolism on HFpEF are also discussed.
Collapse
Affiliation(s)
- Ezra B Ketema
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada. https://twitter.com/Ketema
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
6
|
Mzimela NC, Sosibo AM, Ngubane PS, Khathi A. Investigation into changes in inflammatory and immune cell markers in pre-diabetic patients from Durban, South Africa. J Immunotoxicol 2024; 21:2290282. [PMID: 38099331 DOI: 10.1080/1547691x.2023.2290282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
The prevalence of pre-diabetes is increasing in rapidly urbanizing cities, especially in individuals aged 25 - 45 years old. Studies also indicate that this condition is associated with aberrant immune responses that are also influenced by environmental factors. This study sought to investigate changes in the concentration of immune cells and select inflammatory markers in patients with pre-diabetes in Durban, South Africa. Blood samples collected from King Edward Hospital, after obtaining ethics approval, were divided into non-diabetic (ND), pre-diabetic (PD) and type 2 diabetic (T2D) using ADA criteria. In each sample, the concentration of immune cells and select inflammatory markers were determined. The results showed a significant increase in eosinophil and basophil levels in the PD group as compared to the ND group. Compared to ND, the PD and T2D groups had significant increases in serum TNFα, CD40L and fibrinogen concentrations. Additionally, there were decreases in serum CRP, IL-6, and P-selectin in the PD group while these markers increased in the T2D group. These findings were indicative of immune activation and highlight the impact of pre-diabetes in this population. More studies are recommended with a higher number of samples that are stratified by gender and represent the gender ratio in the city.
Collapse
Affiliation(s)
- Nomusa Christina Mzimela
- School of Laboratory Medicine and Medical Science, College of Health Sciences
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | | | - Andile Khathi
- School of Laboratory Medicine and Medical Science, College of Health Sciences
| |
Collapse
|
7
|
Li Y, Li X, Han Z, Yang R, Zhou W, Peng Y, He J, Liu S. Population structure and selective signature analysis of local sheep breeds in Xinjiang, China based on high-density SNP chip. Sci Rep 2024; 14:28133. [PMID: 39548146 PMCID: PMC11568293 DOI: 10.1038/s41598-024-76573-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
The frigid and droughty climate of Xinjiang in China has given rise to unique indigenous sheep breeds with robust adaptability and resistance. To investigate the genetic mechanism of adaptability of Xinjiang sheep to the local extreme environment, we conducted population genetic structure analyses for three native Xinjiang sheep breeds: Altay sheep (ALT), Bashbay Sheep (BSBC), and Duolang sheep (DLC), as well as two foreign sheep breeds: Suffolk and Dorset, using the Ovine Infinium HD SNP BeadChip(680 K). Our findings revealed distinct genetic and evolutionary histories between Xinjiang and foreign sheep breeds. Principal Component Analysis (PCA) and phylogenetic tree effectively differentiate these five sheep breeds based on their geographical origins, and the domestication level of Xinjiang sheep is comparatively lower than that of foreign sheep breeds. Furthermore, by utilizing three selective signature methods, namely Fixation Index (Fst), Cross Population Extended Haplotype Homozygosity Test (XP-EHH), and Nucleotide Diversity (π), we have successfully identified 22 potential candidate genes. Among these genes, there are TBXT, PDGFD, and VEGFA, which are closely related to tail type and lipid metabolism; VIL1, SLC11A1, and ZBTB46, which are associated with immune function; and candidate genes such as BNC1, HDAC1, and BMP5, which impact sheep reproductive traits. This study establishes a foundation for conserving and utilizing local sheep germplasm resources in Xinjiang and provides molecular insights into the genetic mechanisms governing sheep adaptation to extreme cold and arid environments.
Collapse
Affiliation(s)
- Yanhao Li
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Xiaopeng Li
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Zhipeng Han
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Ruizhi Yang
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
- College of Life Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
| | - Wen Zhou
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Yuwei Peng
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Jianzhong He
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China.
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China.
| | - Shudong Liu
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China.
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China.
| |
Collapse
|
8
|
Elkanawati RY, Sumiwi SA, Levita J. Impact of Lipids on Insulin Resistance: Insights from Human and Animal Studies. Drug Des Devel Ther 2024; 18:3337-3360. [PMID: 39100221 PMCID: PMC11298177 DOI: 10.2147/dddt.s468147] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
Insulin resistance (IR) is a complex pathological condition central to metabolic diseases such as type 2 diabetes mellitus (T2DM), cardiovascular disease, non-alcoholic fatty liver disease, and polycystic ovary syndrome (PCOS). This review evaluates the impact of lipids on insulin resistance (IR) by analyzing findings from human and animal studies. The articles were searched on the PubMed database using two keywords: (1) "Role of Lipids AND Insulin Resistance AND Humans" and (2) "Role of Lipids AND Insulin Resistance AND Animal Models". Studies in humans revealed that elevated levels of free fatty acids (FFAs) and triglycerides (TGs) are closely associated with reduced insulin sensitivity, and interventions like metformin and omega-3 fatty acids show potential benefits. In animal models, high-fat diets disrupt insulin signaling and increase inflammation, with lipid mediators such as diacylglycerol (DAG) and ceramides playing significant roles. DAG activates protein kinase C, which eventually impairs insulin signaling, while ceramides inhibit Akt/PKB, further contributing to IR. Understanding these mechanisms is crucial for developing effective prevention and treatment strategies for IR-related diseases.
Collapse
Affiliation(s)
- Rani Yulifah Elkanawati
- Master Program in Pharmacy, Faculty of Pharmacy, Padjadjaran University, Jawa Barat, West Java, 45363, Indonesia
| | - Sri Adi Sumiwi
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, West Java, 45363, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang, West Java, 45363, Indonesia
| |
Collapse
|
9
|
Li X, Zhou Q. Relationship of weight-adjusted waist index and developmental disabilities in children 6 to 17 years of age: a cross-sectional study. Front Endocrinol (Lausanne) 2024; 15:1406996. [PMID: 39027477 PMCID: PMC11254689 DOI: 10.3389/fendo.2024.1406996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Purpose The development of multiple system diseases is increased by obesity. However, the connection between obesity and developmental disabilities (DDs) in children is unclear. As an obesity index, the weight-adjusted waist index (WWI) assessed fat distribution and muscle mass. In this study, we examined the correlation between WWI and DDs among children 6 to 17 years of age. Methods This study used data from the National Health and Nutrition Examination Survey database (NHANES) covering 2003 to 2018, which included the data of 17,899 participants between 6 and 17 years of age. Data regarding their waist circumference, weight, and DDs were collected via physical examinations and questionnaire, respectively. A person's WWI is calculated by dividing their waist circumference by their weight squared. The correlation between WWI and DDs was studied using weighted multiple logistic regression models. Additionally, a sensitivity analysis was conducted utilizing a generalized additive model and smooth curve fitting. Results After adjusting for all covariates, WWI was positively related to DDs in children ages 6-17. Based on the sensitivity analysis, the correlation between the WWI and prevalence of DDs remained consistent across subgroups. Additionally, there was a J-shaped correlation between the WWI and the prevalence of DDs in children ages 6 through 11. Conclusion Children 6-17 years of age with a high WWI were at greater risk for DDs; however, the causal relationships and potential mechanisms require further exploration.
Collapse
Affiliation(s)
| | - Qi Zhou
- Department of Neonatal, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Luo Y, Luo D, Li M, Tang B. Insulin Resistance in Pediatric Obesity: From Mechanisms to Treatment Strategies. Pediatr Diabetes 2024; 2024:2298306. [PMID: 40302954 PMCID: PMC12016791 DOI: 10.1155/2024/2298306] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/07/2024] [Accepted: 06/15/2024] [Indexed: 05/02/2025] Open
Abstract
Insulin resistance, an increasingly prevalent characteristic among children and adolescents with obesity, is now recognized as a significant contributor to the development of type 2 diabetes mellitus (T2DM) and other metabolic diseases in individuals with obesity. Insulin resistance refers to a decrease in the sensitivity of peripheral tissues (primarily skeletal muscle, adipose tissue, and liver) to insulin, which is mainly characterized by impaired glucose uptake and utilization. Although the mechanisms underlying insulin resistance in children with obesity remain incompletely elucidated, several risk factors including lipid metabolism disorders, oxidative stress (OS), mitochondrial dysfunction, inflammation, and genetic factors have been identified as pivotal contributors to the pathogenesis of obesity-related insulin resistance. In this review, we comprehensively analyze relevant literature and studies to elucidate the underlying mechanisms of insulin resistance in childhood obesity. Additionally, we discuss treatment strategies for pediatric obesity from a perspective centered on improving insulin sensitivity, aiming to provide valuable insights for the prevention and management of pediatric obesity.
Collapse
Affiliation(s)
- Yu Luo
- Department of PediatricsSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of China, Chengdu, China
| | - Dan Luo
- Department of PediatricsSchool of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maojun Li
- Department of PediatricsSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of China, Chengdu, China
| | - Binzhi Tang
- Department of PediatricsSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of China, Chengdu, China
- Department of PediatricsSchool of Medicine and Life Science of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Luo HY, Mu WJ, Chen M, Zhu JY, Li Y, Li S, Yan LJ, Li RY, Yin MT, Li X, Chen HM, Guo L. Hepatic Klf10-Fh1 axis promotes exercise-mediated amelioration of NASH in mice. Metabolism 2024; 155:155916. [PMID: 38615945 DOI: 10.1016/j.metabol.2024.155916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Exercise is an effective non-pharmacological strategy for the treatment of nonalcoholic steatohepatitis (NASH), but the underlying mechanism needs further investigation. Kruppel-like factor 10 (Klf10) is a transcriptional factor that is expressed in multiple tissues including liver, whose role in NASH is not well defined. In our study, exercise induces hepatic Klf10 expression through the cAMP/PKA/CREB pathway. Hepatocyte-specific knockout of Klf10 (Klf10LKO) increases lipid accumulation, cell death, inflammation and fibrosis in NASH diet-fed mice and reduces the protective effects of treadmill exercise against NASH, while hepatocyte-specific overexpression of Klf10 (Klf10LTG) works in concert with exercise to reduce NASH in mice. Mechanistically, Klf10 promotes the expression of fumarate hydratase 1 (Fh1), thereby reducing fumarate accumulation in hepatocytes. This decreases the trimethyl (me3) levels of histone 3 lysine 4 (H3K4me3) on lipogenic genes promoters to attenuate lipogenesis, thus ameliorating free fatty acids (FFAs)-induced hepatocytes steatosis, apoptosis, insulin resistance and blunting dysfunctional hepatocytes-mediated activation of macrophages and hepatic stellate cells. Therefore, by regulating the Fh1/fumarate/H3K4me3 pathway, Klf10 acts as a downstream effector of exercise to combat NASH.
Collapse
Affiliation(s)
- Hong-Yang Luo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Wang-Jing Mu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Yang Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Shan Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Lin-Jing Yan
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Ruo-Ying Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Meng-Ting Yin
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Xin Li
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Hu-Min Chen
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai 200438, China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
12
|
Zhu L, Litts B, Wang Y, Rein JA, Atzrodt CL, Chinnarasu S, An J, Thorson AS, Xu Y, Stafford JM. Ablation of IFNγ in myeloid cells suppresses liver inflammation and fibrogenesis in mice with hepatic small heterodimer partner (SHP) deletion. Mol Metab 2024; 83:101932. [PMID: 38589002 PMCID: PMC11035112 DOI: 10.1016/j.molmet.2024.101932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common complication of obesity and, in severe cases, progresses to metabolic dysfunction-associated steatohepatitis (MASH). Small heterodimer partner (SHP) is an orphan member of the nuclear receptor superfamily and regulates metabolism and inflammation in the liver via a variety of pathways. In this study, we investigate the molecular foundation of MASH progression in mice with hepatic SHP deletion and explore possible therapeutic means to reduce MASH. METHODS Hepatic SHP knockout mice (SHPΔhep) and their wild-type littermates (SHPfl/fl) of both sexes were fed a fructose diet for 14 weeks and subjected to an oral glucose tolerance test. Then, plasma lipids were determined, and liver lipid metabolism and inflammation pathways were analyzed with immunoblotting, RNAseq, and qPCR assays. To explore possible therapeutic intersections of SHP and inflammatory pathways, SHPΔhep mice were reconstituted with bone marrow lacking interferon γ (IFNγ-/-) to suppress inflammation. RESULTS Hepatic deletion of SHP in mice fed a fructose diet decreased liver fat and increased proteins for fatty acid oxidation and liver lipid uptake, including UCP1, CPT1α, ACDAM, and SRBI. Despite lower liver fat, hepatic SHP deletion increased liver inflammatory F4/80+ cells and mRNA levels of inflammatory cytokines (IL-12, IL-6, Ccl2, and IFNγ) in both sexes and elevated endoplasmic reticulum stress markers of Cox2 and CHOP in female mice. Liver bulk RNAseq data showed upregulation of genes whose protein products regulate lipid transport, fatty acid oxidation, and inflammation in SHPΔhep mice. The increased inflammation and fibrosis in SHPΔhep mice were corrected with bone marrow-derived IFNγ-/- myeloid cell transplantation. CONCLUSION Hepatic deletion of SHP improves fatty liver but worsens hepatic inflammation possibly by driving excess fatty acid oxidation, which is corrected by deletion of IFNγ specifically in myeloid cells. This suggests that hepatic SHP limits fatty acid oxidation during fructose diet feeding but, in doing so, prevents pro-MASH pathways. The IFNγ-mediated inflammation in myeloid cells appears to be a potential therapeutic target to suppress MASH.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | - Bridget Litts
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey A Rein
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | | | | | - Julia An
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA
| | - Ariel S Thorson
- Department of Molecular Physiology & Biophysics, Vanderbilt University, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John M Stafford
- Tennessee Valley Health System, Veterans Affairs, Nashville, TN, USA; Department of Molecular Physiology & Biophysics, Vanderbilt University, USA; Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, USA.
| |
Collapse
|
13
|
Lekakis V, Papatheodoridis GV. Natural history of metabolic dysfunction-associated steatotic liver disease. Eur J Intern Med 2024; 122:3-10. [PMID: 37940495 DOI: 10.1016/j.ejim.2023.11.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), which has been the term for non-alcoholic fatty liver disease (NAFLD) since June 2023, represents the most common liver disease worldwide and is a leading cause of liver-related morbidity and mortality. A thorough knowledge of the disease's natural history is required to promptly stratify patients' risks, since MASLD is a multifaceted disorder with a broad range of clinical phenotypes. The histological disease spectrum ranges from isolated hepatic steatosis, currently named as metabolic dysfunction-associated steatotic liver (MASL), to metabolic dysfunction-associated steatohepatitis (MASH) and eventually may accumulate hepatic fibrosis and develop cirrhosis and/or hepatocellular carcinoma (HCC). Several risk factors for fibrosis progression have been identified, while the disease's progression displays notable dynamism and bidirectionality. When compared to the general population, all MASLD histological stages are substantially related with greater overall mortality, and this association exhibits a disease severity-dependent pattern. Interestingly, the fibrosis stage is the most accurate predictor of mortality among MASLD patients. The mortality attributed to MASLD predominantly stems from issues linked with the liver and cardiovascular system, as well as HCC and extrahepatic cancers. In light of the disease natural course, it is crucial to prioritize the identification of at-risk patients for disease progression in order to effectively address and change modifiable risk factors, hence mitigating disease complications. Further investigation is required to define the phenotype of rapid progressors more precisely as well as to improve risk stratification for HCC in non-cirrhotic individuals.
Collapse
Affiliation(s)
- Vasileios Lekakis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma Street, Athens 11527, Greece
| | - George V Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma Street, Athens 11527, Greece.
| |
Collapse
|
14
|
Zhang C, Steadman M, Santos HP, Shaikh SR, Xavier RM. GPAT1 Activity and Abundant Palmitic Acid Impair Insulin Suppression of Hepatic Glucose Production in Primary Mouse Hepatocytes. J Nutr 2024; 154:1109-1118. [PMID: 38354952 PMCID: PMC11007742 DOI: 10.1016/j.tjnut.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Glycerol-3-phosphate acyltransferase (GPAT) activity is correlated with obesity and insulin resistance in mice and humans. However, insulin resistance exists in people with normal body weight, and individuals with obesity may be metabolically healthy, implying the presence of complex pathophysiologic mechanisms underpinning insulin resistance. OBJECTIVE We asked what conditions related to GPAT1 must be met concurrently for hepatic insulin resistance to occur. METHODS Mouse hepatocytes were overexpressed with GPATs via adenoviral infection or exposed to high or low concentrations of glucose. Glucose production by the cells and phosphatidic acid (PA) content in the cells were assayed, GPAT activity was measured, relative messenger RNA expressions of sterol-regulatory element-binding protein 1c (SREBP1c), carbohydrate response element-binding protein (ChREBP), and GPAT1 were analyzed, and insulin signaling transduction was examined. RESULTS Overexpressing GPAT1 in mouse hepatocytes impaired insulin's suppression of glucose production, together with an increase in both N-ethylmaleimide-resistant GPAT activity and the content of di-16:0 PA. Akt-mediated insulin signaling was inhibited in hepatocytes that overexpressed GPAT1. When the cells were exposed to high-glucose concentrations, insulin suppression of glucose production was impaired, and adding palmitic acid exacerbated this impairment. High-glucose exposure increased the expression of SREBP1c, ChREBP, and GPAT1 by ∼2-, 5-, and 5.7-fold, respectively. The addition of 200 mM palmitic acid or linoleic acid to the culture media did not change the upregulation of expression of these genes by high glucose. High-glucose exposure increased di-16:0 PA content in the cells, and adding palmitic acid further increased di-16:0 PA content. The effect was specific to palmitic acid because linoleic acid did not show these effects. CONCLUSION These data demonstrate that high-GPAT1 activity, whether induced by glucose exposure or acquired by transfection, and abundant palmitic acid can impair insulin's ability to suppress hepatic glucose production in primary mouse hepatocytes.
Collapse
Affiliation(s)
- Chongben Zhang
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Mathew Steadman
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Hudson P Santos
- School of Nursing and Health Studies, University of Miami, Coral Gables, FL, United States
| | - Saame R Shaikh
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rose Mary Xavier
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
15
|
Tan J, Xiao A, Yang L, Tao YL, Shao Y, Zhou Q. Diabetes and high-glucose could upregulate the expression of receptor for activated C kinase 1 in retina. World J Diabetes 2024; 15:519-529. [PMID: 38591093 PMCID: PMC10999037 DOI: 10.4239/wjd.v15.i3.519] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/08/2023] [Accepted: 01/18/2024] [Indexed: 03/15/2024] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a major ocular complication of diabetes mellitus, leading to visual impairment. Retinal pigment epithelium (RPE) injury is a key component of the outer blood retinal barrier, and its damage is an important indicator of DR. Receptor for activated C kinase 1 (RACK1) activates protein kinase C-ε (PKC-ε) to promote the generation of reactive oxygen species (ROS) in RPE cells, leading to apoptosis. Therefore, we hypothesize that the activation of RACK1 under hypoxic/high-glucose conditions may promote RPE cell apoptosis by modulating PKC-ε/ROS, thereby disrupting the barrier effect of the outer blood retinal barrier and contributing to the progression of DR. AIM To investigate the role and associated underlying mechanisms of RACK1 in the development of early DR. METHODS In this study, Sprague-Dawley rats and adult RPE cell line-19 (ARPE-19) cells were used as in vivo and in vitro models, respectively, to explore the role of RACK1 in mediating PKC-ε in early DR. Furthermore, the impact of RACK1 on apoptosis and barrier function of RPE cells was also investigated in the former model. RESULTS Streptozotocin-induced diabetic rats showed increased apoptosis and up-regulated expression of RACK1 and PKC-ε proteins in RPE cells following a prolonged modeling. Similarly, ARPE-19 cells exposed to high glucose and hypoxia displayed elevated mRNA and protein levels of RACK1 and PKC-ε, accompanied by an increases in ROS production, apoptosis rate, and monolayer permeability. However, silencing RACK1 significantly downregulated the expression of PKC-ε and ROS, reduced cell apoptosis and permeability, and protected barrier function. CONCLUSION RACK1 plays a significant role in the development of early DR and might serve as a potential therapeutic target for DR by regulating RPE apoptosis and barrier function.
Collapse
Affiliation(s)
- Jian Tan
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ang Xiao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Lin Yang
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yu-Lin Tao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
16
|
Yuan L, Verhoeven A, Blomberg N, van Eyk HJ, Bizino MB, Rensen PCN, Jazet IM, Lamb HJ, Rabelink TJ, Giera M, van den Berg BM. Ethnic Disparities in Lipid Metabolism and Clinical Outcomes between Dutch South Asians and Dutch White Caucasians with Type 2 Diabetes Mellitus. Metabolites 2024; 14:33. [PMID: 38248836 PMCID: PMC10819672 DOI: 10.3390/metabo14010033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/26/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) poses a higher risk for complications in South Asian individuals compared to other ethnic groups. To shed light on potential mediating factors, we investigated lipidomic changes in plasma of Dutch South Asians (DSA) and Dutch white Caucasians (DwC) with and without T2DM and explore their associations with clinical features. Using a targeted quantitative lipidomics platform, monitoring over 1000 lipids across 17 classes, along with 1H NMR based lipoprotein analysis, we studied 51 healthy participants (21 DSA, 30 DwC) and 92 T2DM patients (47 DSA, 45 DwC) from the MAGNetic resonance Assessment of VICTOza efficacy in the Regression of cardiovascular dysfunction in type 2 dIAbetes mellitus (MAGNA VICTORIA) study. This comprehensive mapping of the circulating lipidome allowed us to identify relevant lipid modules through unbiased weighted correlation network analysis, as well as disease and ethnicity related key mediatory lipids. Significant differences in lipidomic profiles, encompassing various lipid classes and species, were observed between T2DM patients and healthy controls in both the DSA and DwC populations. Our analyses revealed that healthy DSA, but not DwC, controls already exhibited a lipid profile prone to develop T2DM. Particularly, in DSA-T2DM patients, specific lipid changes correlated with clinical features, particularly diacylglycerols (DGs), showing significant associations with glycemic control and renal function. Our findings highlight an ethnic distinction in lipid modules influencing clinical outcomes in renal health. We discover distinctive ethnic disparities of the circulating lipidome and identify ethnicity-specific lipid markers. Jointly, our discoveries show great potential as personalized biomarkers for the assessment of glycemic control and renal function in DSA-T2DM individuals.
Collapse
Affiliation(s)
- Lushun Yuan
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.Y.); (P.C.N.R.); (T.J.R.)
- Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.V.); (N.B.); (M.G.)
| | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.V.); (N.B.); (M.G.)
| | - Huub J. van Eyk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (H.J.v.E.); (I.M.J.)
| | - Maurice B. Bizino
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.B.B.); (H.J.L.)
| | - Patrick C. N. Rensen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.Y.); (P.C.N.R.); (T.J.R.)
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (H.J.v.E.); (I.M.J.)
| | - Ingrid M. Jazet
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (H.J.v.E.); (I.M.J.)
| | - Hildo J. Lamb
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.B.B.); (H.J.L.)
| | - Ton J. Rabelink
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.Y.); (P.C.N.R.); (T.J.R.)
- Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (A.V.); (N.B.); (M.G.)
| | - Bernard M. van den Berg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (L.Y.); (P.C.N.R.); (T.J.R.)
- Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
17
|
Syed-Abdul MM. Lipid Metabolism in Metabolic-Associated Steatotic Liver Disease (MASLD). Metabolites 2023; 14:12. [PMID: 38248815 PMCID: PMC10818604 DOI: 10.3390/metabo14010012] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Metabolic-associated steatotic liver disease (MASLD) is a cluster of pathological conditions primarily developed due to the accumulation of ectopic fat in the hepatocytes. During the severe form of the disease, i.e., metabolic-associated steatohepatitis (MASH), accumulated lipids promote lipotoxicity, resulting in cellular inflammation, oxidative stress, and hepatocellular ballooning. If left untreated, the advanced form of the disease progresses to fibrosis of the tissue, resulting in irreversible hepatic cirrhosis or the development of hepatocellular carcinoma. Although numerous mechanisms have been identified as significant contributors to the development and advancement of MASLD, altered lipid metabolism continues to stand out as a major factor contributing to the disease. This paper briefly discusses the dysregulation in lipid metabolism during various stages of MASLD.
Collapse
Affiliation(s)
- Majid Mufaqam Syed-Abdul
- Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
18
|
Staltner R, Burger K, Baumann A, Bergheim I. Fructose: a modulator of intestinal barrier function and hepatic health? Eur J Nutr 2023; 62:3113-3124. [PMID: 37596353 PMCID: PMC10611622 DOI: 10.1007/s00394-023-03232-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023]
Abstract
PURPOSE Consumption of fructose has repeatedly been discussed to be a key factor in the development of health disturbances such as hypertension, diabetes type 2, and non-alcoholic fatty liver disease. Despite intense research efforts, the question if and how high dietary fructose intake interferes with human health has not yet been fully answered. RESULTS Studies suggest that besides its insulin-independent metabolism dietary fructose may also impact intestinal homeostasis and barrier function. Indeed, it has been suggested by the results of human and animal as well as in vitro studies that fructose enriched diets may alter intestinal microbiota composition. Furthermore, studies have also shown that both acute and chronic intake of fructose may lead to an increased formation of nitric oxide and a loss of tight junction proteins in small intestinal tissue. These alterations have been related to an increased translocation of pathogen-associated molecular patterns (PAMPs) like bacterial endotoxin and an induction of dependent signaling cascades in the liver but also other tissues. CONCLUSION In the present narrative review, results of studies assessing the effects of fructose on intestinal barrier function and their impact on the development of health disturbances with a particular focus on the liver are summarized and discussed.
Collapse
Affiliation(s)
- Raphaela Staltner
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Katharina Burger
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Anja Baumann
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria.
| |
Collapse
|
19
|
Zhou X, Zhang J, Shen J, Cheng B, Bi C, Ma Q. Branched-chain amino acid modulation of lipid metabolism, gluconeogenesis, and inflammation in a finishing pig model: targeting leucine and valine. Food Funct 2023; 14:10119-10134. [PMID: 37882496 DOI: 10.1039/d3fo03899h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Branched-chain amino acids (BCAAs) play a regulatory role in adipogenesis and energy balance. Therefore, this study aimed to investigate the impact of BCAA supplements, especially leucine (Leu) and valine (Val) supplementation, on lipid metabolism and related disorders in a finishing pig model. The results demonstrated that Leu (1%) and Val decreased serum as well as hepatic lipid accumulation. Moreover, metabolomics and lipidomics analyses revealed that Leu and Val markedly downregulated the level of various lipid species in the liver. This outcome may be explained by Leu and Val promoting cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)/hormone-sensitive triglyceride lipase (HSL) signaling pathways. Leu and Val altered the fatty acid composition in distinct adipose tissues and decreased the levels of inflammatory factors. Additionally, they significantly decreased back fat thickness, and the results of the fatty acid profiles demonstrated that Leu and Val significantly increased the levels of monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs) while decreasing those of saturated fatty acids (SFAs), especially in back fat and abdominal fat. Besides, Leu and Val restored glucose homeostasis by suppressing gluconeogenesis through the serine/threonine protein kinase (AKT)/transcription factor forkhead box O1 (FOXO1) signaling pathway in the liver and back fat. In summary, these results suggest that Leu and Val may serve as key regulators for modulating lipid metabolism and steatosis.
Collapse
Affiliation(s)
- Xinbo Zhou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Junjie Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Jian Shen
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Baojing Cheng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Chongpeng Bi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Qingquan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
20
|
El-Amouri S, Karakashian A, Bieberich E, Nikolova-Karakashian M. Regulated translocation of neutral sphingomyelinase-2 to the plasma membrane drives insulin resistance in steatotic hepatocytes. J Lipid Res 2023; 64:100435. [PMID: 37640282 PMCID: PMC10550728 DOI: 10.1016/j.jlr.2023.100435] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Obesity-associated diabetes is linked to the accumulation of ceramide in various organs, including the liver. The exact mechanisms by which ceramide contributes to diabetic pathology are unclear, but one proposed scenario is that ceramide accumulation may inhibit insulin signaling pathways. It is unknown however whether the excess ceramide is generated proximal to the insulin receptor, that is, at the plasma membrane (PM), where it could affect the insulin signaling pathway directly, or the onset of insulin resistance is due to ceramide-induced mitochondrial dysfunction and/or lipotoxicity. Using hepatic cell lines and primary cultures, gain- and loss- of function approach, and state-of-the art lipid imaging, this study shows that PM-associated neutral sphingomyelinase 2 (nSMase2) regulates ceramide homeostasis in fat-loaded hepatocytes and drives the onset of insulin resistance. Our results provide evidence of a regulated translocation of nSMase2 to the PM which leads to local generation of ceramide and insulin resistance in cells treated with palmitic acid (PAL), a type of fat commonly found in diabetogenic diets. Oleic acid, which also causes accumulation of lipid droplets, does not induce nSMase2 translocation and insulin resistance. Experiments using the acyl-biotin exchange method to quantify protein palmitoylation show that cellular PAL abundance regulates the rate of nSMase2 palmitoylation. Furthermore, while inhibition of nSMase2 with GW4869 prevents PAL-induced insulin resistance, the overexpression of wild type nSMase2 but not palmitoylation-defective mutant protein potentiates the suppressive effect of PAL on insulin signaling. Overall, this study identifies nSMase2 as a novel component of the mechanism of insulin resistance onset in fat-loaded hepatocytes, that is, cell-autonomous and driven by PAL.
Collapse
Affiliation(s)
- S El-Amouri
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - A Karakashian
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - E Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - M Nikolova-Karakashian
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
21
|
Xiao J, Zhang X, Zhu C, Gu Y, Sun L, Liang X, He Q. Development, Validation, and Application of a Scoring Model for Non-alcoholic Steatohepatitis. Obes Surg 2023; 33:3246-3255. [PMID: 37644345 DOI: 10.1007/s11695-023-06804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 08/12/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE The aim of this study was to explore risk factors of NASH and then develop a non-invasive scoring model in Chinese patients with obesity. A scoring system was then applied to assess the effect of sleeve gastrectomy on NASH. METHODS A total of 243 patients with obesity were included and divided into NASH group and non-NASH group according to the pathological results of liver biopsy. Logistic regression was used to determine risk factors of NASH. A scoring model was derived by risk factors of NASH. Then, postoperative follow-up was performed in 70 patients. RESULTS Among the 243 patients, 118 (48.56%) patients showed NASH. Multivariate logistic regression identified aspartate aminotransferase (AST) (>21.50 IU/L), high-density lipoprotein cholesterol (HDL-C) (<1.155mmol/L), and homeostasis model assessment (HOMA-IR) (>9.368) as independent risk factors of NASH. The model included above risk factors showed a negative predictive value (NPV) of 70.38% in the low-risk category and a positive predictive value (PPV) of 85.71% in the high-risk category, with the area under the receiver operator curve (AUROC) of 0.737. Bariatric surgery resulted in a sharp decline in AST and HOMA-IR and a significant increase of HDL-C. The points of scoring model were improved at 6 months after surgery. CONCLUSION A non-invasive scoring model was derived by the risk factors of NASH included AST, HDL-C, and HOMA-IR and applied to the postoperative follow-up. After sleeve gastrectomy, the above risk factors and points of scoring model were significantly improved.
Collapse
Affiliation(s)
- Jinfeng Xiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xinxin Zhang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chonggui Zhu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yian Gu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoyu Liang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
22
|
Rath S, Maiti D, Modi M, Pal P, Munan S, Mohanty B, Bhatia A, Bhowal R, Priyadarshini R, Samanta A, Munshi P, Sen S. Metal-free synthesis and study of glycine betaine derivatives in water for antimicrobial and anticancer applications. iScience 2023; 26:107285. [PMID: 37575199 PMCID: PMC10415718 DOI: 10.1016/j.isci.2023.107285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/30/2023] [Indexed: 08/15/2023] Open
Abstract
A sustainable synthesis of interesting glycine betaine derivatives from cyclic 3°-amines viz. N-methyl morpholine (NMM), N-methyl piperidine (NMP), and 1,4-diazabicyclo[2.2.2]octane (DABCO) with numerous aryl diazoacetates 1 in water and under blue LED is reported. Generally, 3°-amines and metal carbenoids (from diazoacetates with transition metal catalysts) provide C-H insertion at the α-position of the amines. Computational comparison of the metal carbenoid with the singlet carbene (metal free and generated under blue LED) realized the difference in reactivity. Next, experimental results corroborated the preliminary findings. The products were isolated either by precipitation of the solid or gel-like final products from the aqueous reaction mixture without any chromatographic purification. The reaction mechanism was realized by control experiments. These compounds exhibit selective bactericidal properties against Gram-positive S. aureus, induce lipid droplets (LDs) formation in HePG2 cells and single crystal X-ray diffraction study of their halogenated analogs reveal interesting Hal … Hal contacts.
Collapse
Affiliation(s)
- Suchismita Rath
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Debajit Maiti
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Malvika Modi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Parul Pal
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Subrata Munan
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Biswajit Mohanty
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Anjani Bhatia
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Rohit Bhowal
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Richa Priyadarshini
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Animesh Samanta
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Parthapratim Munshi
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| | - Subhabrata Sen
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Gautam Buddha Nagar, Chithera, Dadri, UP 201310, India
| |
Collapse
|
23
|
Errafii K, Boujraf S, Chikri M. Transcriptomic Analysis from Normal Glucose Tolerance to T2D of Obese Individuals Using Bioinformatic Tools. Int J Mol Sci 2023; 24:ijms24076337. [PMID: 37047308 PMCID: PMC10093815 DOI: 10.3390/ijms24076337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Understanding the role of white adipose tissue (WAT) in the occurrence and progression of metabolic syndrome is of considerable interest; among the metabolic syndromes are obesity and type 2 diabetes (T2D). Insulin resistance is a key factor in the development of T2D. When the target cells become resistant to insulin, the pancreas responds by producing more insulin to try to lower blood glucose. Over time, this can lead to a state of hyperinsulinemia (high levels of insulin in the blood), which can further exacerbate insulin resistance and contribute to the development of T2D. In order to understand the difference between healthy and unhealthy obese individuals, we have used published transcriptomic profiling to compare differences between the WAT obtained from obese diabetics and subjects who are obese with normal glucose tolerance and insulin resistance. The identification of aberrantly expressed messenger RNA (mRNA) and the resulting molecular interactions and signaling networks is essential for a better understanding of the progression from normal glucose-tolerant obese individuals to obese diabetics. Computational analyses using Ingenuity Pathway Analysis (IPA) identified multiple activated signaling networks in obesity progression from insulin-resistant and normal glucose-tolerant (IR-NGT) individuals to those with T2D. The pathways affected are: Tumor Necrosis Factor (TNF), Extracellular signal-Regulated protein Kinase 1/2 ERK1/2, Interleukin 1 A (IL1A), Protein kinase C (Pkcs), Convertase C5, Vascular endothelial growth factor (Vegf), REL-associated protein (RELA), Interleukin1/1 B (IL1/1B), Triggering receptor expressed on myeloid cells (TREM1) and Nuclear factor KB1 (NFKB1) networks, while functional annotation highlighted Liver X Receptor (LXR) activation, phagosome formation, tumor microenvironment pathway, LPS/IL-1 mediated inhibition of RXR function, TREM1 signaling and IL-6 signaling. Together, by conducting a thorough bioinformatics study of protein-coding RNAs, prospective targets could be exploited to clarify the molecular pathways underlying the development of obesity-related type 2 diabetes.
Collapse
Affiliation(s)
- Khaoula Errafii
- Clinical Neurosciences Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohammad Ben Abdullah University, Fez 30000, Morocco
- Biochemistry and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohammad Ben Abdullah University, Fez 30000, Morocco
- African Genome Center, Mohamed IV Polytechnic University, Benguerir 43151, Morocco
| | - Said Boujraf
- Clinical Neurosciences Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohammad Ben Abdullah University, Fez 30000, Morocco
- Biochemistry and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohammad Ben Abdullah University, Fez 30000, Morocco
| | - Mohamed Chikri
- Clinical Neurosciences Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohammad Ben Abdullah University, Fez 30000, Morocco
- Biochemistry and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohammad Ben Abdullah University, Fez 30000, Morocco
- Inserm Unite CNRS, Lille University UMR 1283-8199, F-59000 Lille, France
- Correspondence:
| |
Collapse
|
24
|
Gunasekar SK, Heebink J, Carpenter DH, Kumar A, Xie L, Zhang H, Schilling JD, Sah R. Adipose-targeted SWELL1 deletion exacerbates obesity- and age-related nonalcoholic fatty liver disease. JCI Insight 2023; 8:e154940. [PMID: 36749637 PMCID: PMC10077479 DOI: 10.1172/jci.insight.154940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Healthy expansion of adipose tissue is critical for the maintenance of metabolic health, providing an optimized reservoir for energy storage in the form of triacylglycerol-rich lipoproteins. Dysfunctional adipocytes that are unable to efficiently store lipid can result in lipodystrophy and contribute to nonalcoholic fatty liver disease (NAFLD) and metabolic syndrome. Leucine-rich repeat containing protein 8a/SWELL1 functionally encodes the volume-regulated anion channel complex in adipocytes, is induced in early obesity, and is required for normal adipocyte expansion during high-fat feeding. Adipose-specific SWELL1 ablation (Adipo KO) leads to insulin resistance and hyperglycemia during caloric excess, both of which are associated with NAFLD. Here, we show that Adipo-KO mice exhibited impaired adipose depot expansion and excess lipolysis when raised on a variety of high-fat diets, resulting in increased diacylglycerides and hepatic steatosis, thereby driving liver injury. Liver lipidomic analysis revealed increases in oleic acid-containing hepatic triacylglycerides and injurious hepatic diacylglyceride species, with reductions in hepatocyte-protective phospholipids and antiinflammatory free fatty acids. Aged Adipo-KO mice developed hepatic steatosis on a regular chow diet, and Adipo-KO male mice developed spontaneous, aggressive hepatocellular carcinomas (HCCs). These data highlight the importance of adipocyte SWELL1 for healthy adipocyte expansion to protect against NAFLD and HCC in the setting of overnutrition and with aging.
Collapse
Affiliation(s)
- Susheel K. Gunasekar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Heebink
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Danielle H. Carpenter
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Ashutosh Kumar
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Litao Xie
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Haixia Zhang
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joel D. Schilling
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Sinha RA. Autophagy: A Cellular Guardian against Hepatic Lipotoxicity. Genes (Basel) 2023; 14:553. [PMID: 36874473 PMCID: PMC7614268 DOI: 10.3390/genes14030553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Lipotoxicity is a phenomenon of lipid-induced cellular injury in nonadipose tissue. Excess of free saturated fatty acids (SFAs) contributes to hepatic injury in nonalcoholic fatty liver disease (NAFLD), which has been growing at an unprecedented rate in recent years. SFAs and their derivatives such as ceramides and membrane phospholipids have been shown to induce intrahepatic oxidative damage and ER stress. Autophagy represents a cellular housekeeping mechanism to counter the perturbation in organelle function and activation of stress signals within the cell. Several aspects of autophagy, including lipid droplet assembly, lipophagy, mitophagy, redox signaling and ER-phagy, play a critical role in mounting a strong defense against lipotoxic lipid species within the hepatic cells. This review provides a succinct overview of our current understanding of autophagy-lipotoxicity interaction and its pharmacological and nonpharmacological modulation in treating NAFLD.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
26
|
Zheng ZG, Xu YY, Liu WP, Zhang Y, Zhang C, Liu HL, Zhang XY, Liu RZ, Zhang YP, Shi MY, Yang H, Li P. Discovery of a potent allosteric activator of DGKQ that ameliorates obesity-induced insulin resistance via the sn-1,2-DAG-PKCε signaling axis. Cell Metab 2023; 35:101-117.e11. [PMID: 36525963 DOI: 10.1016/j.cmet.2022.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 09/07/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
sn-1,2-diacylglycerol (sn-1,2-DAG)-mediated activation of protein kinase Cε (PKCε) is a key pathway that is responsible for obesity-related lipid metabolism disorders, which induces hepatic insulin resistance and type 2 diabetes. No small molecules have been previously reported to ameliorate these diseases through this pathway. Here, we screened and identified the phytochemical atractylenolide II (AT II) that reduces the hepatic sn-1,2-DAG levels, deactivates PKCε activity, and improves obesity-induced hyperlipidemia, hepatosteatosis, and insulin resistance. Furthermore, using the ABPP strategy, the diacylglycerol kinase family member DGKQ was identified as a direct target of AT II. AT II may act on a novel drug-binding pocket in the CRD and PH domains of DGKQ to thereby allosterically regulate its kinase activity. Moreover, AT II also increases weight loss by activating DGKQ-AMPK-PGC1α-UCP-1 signaling in adipose tissue. These findings suggest that AT II is a promising lead compound to improve obesity-induced insulin resistance.
Collapse
Affiliation(s)
- Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Yin-Yue Xu
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Wen-Ping Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Yang Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Chong Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Han-Ling Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Xiao-Yu Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Run-Zhou Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Yi-Ping Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Meng-Ying Shi
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hua Yang
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
27
|
Maternal Fructose Intake, Programmed Mitochondrial Function and Predisposition to Adult Disease. Int J Mol Sci 2022; 23:ijms232012215. [DOI: 10.3390/ijms232012215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Fructose consumption is now recognised as a major risk factor in the development of metabolic diseases, such as hyperlipidaemia, diabetes, non-alcoholic fatty liver disease and obesity. In addition to environmental, social, and genetic factors, an unfavourable intrauterine environment is now also recognised as an important factor in the progression of, or susceptibility to, metabolic disease during adulthood. Developmental trajectory in the short term, in response to nutrient restriction or excessive nutrient availability, may promote adaptation that serves to maintain organ functionality necessary for immediate survival and foetal development. Consequently, this may lead to decreased function of organ systems when presented with an unfavourable neonatal, adolescent and/or adult nutritional environment. These early events may exacerbate susceptibility to later-life disease since sub-optimal maternal nutrition increases the risk of non-communicable diseases (NCDs) in future generations. Earlier dietary interventions, implemented in pregnant mothers or those considering pregnancy, may have added benefit. Although, the mechanisms by which maternal diets high in fructose and the vertical transmission of maternal metabolic phenotype may lead to the predisposition to adult disease are poorly understood. In this review, we will discuss the potential contribution of excessive fructose intake during pregnancy and how this may lead to developmental reprogramming of mitochondrial function and predisposition to metabolic disease in offspring.
Collapse
|
28
|
Alshehade S, Alshawsh MA, Murugaiyah V, Asif M, Alshehade O, Almoustafa H, Al Zarzour RH. The role of protein kinases as key drivers of metabolic dysfunction-associated fatty liver disease progression: New insights and future directions. Life Sci 2022; 305:120732. [PMID: 35760093 DOI: 10.1016/j.lfs.2022.120732] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD), proposed in 2020 is a novel term for non-alcoholic fatty liver disease (NAFLD) which was coined for the first time in 1980. It is a leading cause of the most chronic liver disease and hepatic failure all over the world, and unfortunately, with no licensed drugs for treatment yet. The progress of the disease is driven by the triggered inflammatory process, oxidative stress, and insulin resistance in many pathways, starting with simple hepatic steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and liver cancer. Protein kinases (PKs), such as MAPK, ErbB, PKC, PI3K/Akt, and mTOR, govern most of the pathological pathways by acting on various downstream key points in MAFLD and regulating both hepatic gluco- lipo-neogenesis and inflammation. Therefore, modulating the function of those potential protein kinases that are effectively involved in MAFLD might be a promising therapeutic approach for tackling this disease. In the current review, we have discussed the key role of protein kinases in the pathogenesis of MAFLD and performed a protein-protein interaction (PPI) network among the main proteins of each kinase pathway with MAFLD-related proteins to predict the most likely targets of the PKs in MAFLD. Moreover, we have reported the experimental, pre-clinical, and clinical data for the most recent investigated molecules that are activating p38-MAPK and AMPK proteins and inhibiting the other PKs to improve MAFLD condition by regulating oxidation and inflammation signalling.
Collapse
Affiliation(s)
- Salah Alshehade
- Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Vikneswaran Murugaiyah
- Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Muhammad Asif
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Punjab, Pakistan
| | - Omayma Alshehade
- Department of Paediatrics, Faculty of Medicine, Damascus University, Damascus, Syria
| | - Hassan Almoustafa
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Raghdaa Hamdan Al Zarzour
- Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia; Department of Pharmacology, Faculty of Pharmacy, Arab International University, Damascus, Syria.
| |
Collapse
|
29
|
Pan D, Xu L, Guo M. The role of protein kinase C in diabetic microvascular complications. Front Endocrinol (Lausanne) 2022; 13:973058. [PMID: 36060954 PMCID: PMC9433088 DOI: 10.3389/fendo.2022.973058] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Protein kinase C (PKC) is a family of serine/threonine protein kinases, the activation of which plays an important role in the development of diabetic microvascular complications. The activation of PKC under high-glucose conditions stimulates redox reactions and leads to an accumulation of redox stress. As a result, various types of cells in the microvasculature are influenced, leading to changes in blood flow, microvascular permeability, extracellular matrix accumulation, basement thickening and angiogenesis. Structural and functional disorders further exacerbate diabetic microvascular complications. Here, we review the roles of PKC in the development of diabetic microvascular complications, presenting evidence from experiments and clinical trials.
Collapse
Affiliation(s)
- Deng Pan
- Xiyuan hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Lin Xu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ming Guo
- Xiyuan hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
30
|
Shitole SG, Biggs ML, Ix JH, Fretts AM, Tracy RP, Siscovick DS, Djoussé L, Mukamal KJ, Kizer JR. Fasting and Postload Nonesterified Fatty Acids and Glucose Dysregulation in Older Adults. Am J Epidemiol 2022; 191:1235-1247. [PMID: 35247051 PMCID: PMC9989335 DOI: 10.1093/aje/kwac044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 02/07/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
To evaluate the association of nonesterified fatty acids (NEFA) with dysglycemia in older adults, NEFA levels were measured among participants in the Cardiovascular Health Study (United States; enrolled 1989-1993). Associations with insulin sensitivity and pancreatic β-cell function, and with incident type 2 diabetes mellitus (DM), were examined. The sample comprised 2,144 participants (aged 77.9 (standard deviation, 4.5) years). Participant data from the Cardiovascular Health Study visit in 1996-1997 was used with prospective follow-up through 2010. Fasting and postload NEFA showed significant associations with lower insulin sensitivity and pancreatic β-cell function, individually and on concurrent adjustment. Over median follow-up of 9.7 years, 236 cases of DM occurred. Postload NEFA were associated with risk of DM (per standard deviation, hazard ratio = 1.18, 95% confidence interval: 1.08, 1.29), but fasting NEFA were not (hazard ratio = 1.12, 95% confidence interval: 0.97, 1.29). The association for postload NEFA persisted after adjustment for putative intermediates, and after adjustment for fasting NEFA. Sex and body mass index modified these associations, which were stronger for fasting NEFA with DM in men but were accentuated for postload NEFA in women and among leaner individuals. Fasting and postload NEFA were related to lower insulin sensitivity and pancreatic β-cell function, but only postload NEFA were associated with increased DM. Additional study into NEFA metabolism could uncover novel potential targets for diabetes prevention in elders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jorge R Kizer
- Correspondence to Dr. Jorge R. Kizer, 4150 Clement Street, San Francisco, CA 94121 (e-mail: )
| |
Collapse
|
31
|
The Role of Insulin Resistance in Fueling NAFLD Pathogenesis: From Molecular Mechanisms to Clinical Implications. J Clin Med 2022; 11:jcm11133649. [PMID: 35806934 PMCID: PMC9267803 DOI: 10.3390/jcm11133649] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a predominant hepatopathy that is rapidly becoming the most common cause of hepatocellular carcinoma worldwide. The close association with metabolic syndrome’s extrahepatic components has suggested the nature of the systemic metabolic-related disorder based on the interplay between genetic, nutritional, and environmental factors, creating a complex network of yet-unclarified pathogenetic mechanisms in which the role of insulin resistance (IR) could be crucial. This review detailed the clinical and pathogenetic evidence involved in the NAFLD–IR relationship, presenting both the classic and more innovative models. In particular, we focused on the reciprocal effects of IR, oxidative stress, and systemic inflammation on insulin-sensitivity disruption in critical regions such as the hepatic and the adipose tissue, while considering the impact of genetics/epigenetics on the regulation of IR mechanisms as well as nutrients on specific insulin-related gene expression (nutrigenetics and nutrigenomics). In addition, we discussed the emerging capability of the gut microbiota to interfere with physiological signaling of the hormonal pathways responsible for maintaining metabolic homeostasis and by inducing an abnormal activation of the immune system. The translation of these novel findings into clinical practice could promote the expansion of accurate diagnostic/prognostic stratification tools and tailored pharmacological approaches.
Collapse
|
32
|
Lee S, Usman TO, Yamauchi J, Chhetri G, Wang X, Coudriet GM, Zhu C, Gao J, McConnell R, Krantz K, Rajasundaram D, Singh S, Piganelli J, Ostrowska A, Soto-Gutierrez A, Monga SP, Singhi AD, Muzumdar RH, Tsung A, Dong HH. Myeloid FoxO1 depletion attenuates hepatic inflammation and prevents nonalcoholic steatohepatitis. J Clin Invest 2022; 132:154333. [PMID: 35700043 PMCID: PMC9282937 DOI: 10.1172/jci154333] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatic inflammation is culpable for the evolution of asymptomatic steatosis to nonalcoholic steatohepatitis (NASH). Hepatic inflammation results from abnormal macrophage activation. We found that FoxO1 links overnutrition to hepatic inflammation by regulating macrophage polarization and activation. FoxO1 was upregulated in hepatic macrophages, correlating with hepatic inflammation, steatosis and fibrosis in mice and patients with NASH. Myeloid cell-conditional FoxO1 knockout skewed macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 phenotypes, accompanied by the reduction of macrophage infiltration in liver. These effects mitigated overnutrition-induced hepatic inflammation and insulin resistance, contributing to improved hepatic metabolism and increased energy expenditure in myeloid cell FoxO1 knockout mice on HFD. When fed a NASH-inducing diet, myeloid cell FoxO1 knockout mice were protected from developing NASH, culminating in the reduction of hepatic inflammation, steatosis and fibrosis. Mechanistically, FoxO1 counteracts Stat6 to skew macrophage polarization from M2 toward M1 signatures to perpetuate hepatic inflammation in NASH. FoxO1 appears as a pivotal mediator of macrophage activation in response to overnutrition and a therapeutic target for ameliorating hepatic inflammation to stem the disease progression from benign steatosis to NASH.
Collapse
Affiliation(s)
- Sojin Lee
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Taofeek O Usman
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jun Yamauchi
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Goma Chhetri
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Xingchun Wang
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Gina M Coudriet
- Department of Surgery, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Cuiling Zhu
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jingyang Gao
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Riley McConnell
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Kyler Krantz
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Sucha Singh
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jon Piganelli
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Alina Ostrowska
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Radhika H Muzumdar
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Allan Tsung
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, United States of America
| | - H Henry Dong
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| |
Collapse
|
33
|
Identification of the Potential Molecular Mechanisms Linking RUNX1 Activity with Nonalcoholic Fatty Liver Disease, by Means of Systems Biology. Biomedicines 2022; 10:biomedicines10061315. [PMID: 35740337 PMCID: PMC9219880 DOI: 10.3390/biomedicines10061315] [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: 04/13/2022] [Revised: 05/24/2022] [Accepted: 06/01/2022] [Indexed: 12/10/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic hepatic disease; nevertheless, no definitive diagnostic method exists yet, apart from invasive liver biopsy, and nor is there a specific approved treatment. Runt-related transcription factor 1 (RUNX1) plays a major role in angiogenesis and inflammation; however, its link with NAFLD is unclear as controversial results have been reported. Thus, the objective of this work was to determine the proteins involved in the molecular mechanisms between RUNX1 and NAFLD, by means of systems biology. First, a mathematical model that simulates NAFLD pathophysiology was generated by analyzing Anaxomics databases and reviewing available scientific literature. Artificial neural networks established NAFLD pathophysiological processes functionally related to RUNX1: hepatic insulin resistance, lipotoxicity, and hepatic injury-liver fibrosis. Our study indicated that RUNX1 might have a high relationship with hepatic injury-liver fibrosis, and a medium relationship with lipotoxicity and insulin resistance motives. Additionally, we found five RUNX1-regulated proteins with a direct involvement in NAFLD motives, which were NFκB1, NFκB2, TNF, ADIPOQ, and IL-6. In conclusion, we suggested a relationship between RUNX1 and NAFLD since RUNX1 seems to regulate NAFLD molecular pathways, posing it as a potential therapeutic target of NAFLD, although more studies in this field are needed.
Collapse
|
34
|
Inhibition of palmitic acid induced adipogenesis by natural polyphenols in 3T3-L1 adipocytes. In Vitro Cell Dev Biol Anim 2022; 58:396-407. [PMID: 35678984 DOI: 10.1007/s11626-022-00689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/30/2022] [Indexed: 11/05/2022]
Abstract
Dietary free fatty acids induce preadipocyte differentiation in the presence of a hormonal cocktail in 3T3-L1 adipocytes. Plant polyphenols are curb adipocyte differentiation and protect from metabolic stress. In the present study, we examined the effects of the saturated fatty acid, palmitic acid (PA) in presence of flavonoids, chrysin (CR) and hesperidin (HD) and phenolic acid, syringic acid (SYA) and sinapic acid (SIA). Adipocytes were incubated for 10 d with 100 μmol of PA along with 10-100 µmol CR/HD and 100-1000 µmol SYA/SIA. PA induced clonal expansion of preadipocytes, differentiation and oxidative stress in 3T3-L1 cells following 10 d of differentiation. Adipocytes treated with PA exhibited an increase of 300% in clonal population, 110% lipid and 172% reactive oxygen species accumulation. But treatment with CR, HD, SYA and SIA in the presence of PA concentration-dependent effect was observed. Concentrations of CR/HD and SYA/SIA inhibited PA-induced mRNA expression of PPARγ, C/EBPα, SREBP-1c, FAS and NOX4. Moreover, CR, HD, SYA and SIA did not exhibit toxicity in Drosophila DNA. In summary, these results suggest that dietary fatty acids act directly on adipocytes and addition of CR, HD, SYA and SIA resulted in reduction of PA-induced negative effects on 3T3-L1 adipocytes. HIGHLIGHTS: • Palmitic acid, the common dietary free fatty acid, is known to induce adipogenesis in 3T3-L1 adipocytes. • Treatment of differentiating adipocytes with flavonoids and phenolic acids reduced palmitic acid-induced clonal expansion of preadipocytes. • Phytocompounds reduced lipid accumulation and triglyceride production as well as ROS accumulation. • Thus, the phytocompounds showed effective anti-adipogenic activity even in palmitic acid challenged environment in adipocytes.
Collapse
|
35
|
Caturano A, Vetrano E, Galiero R, Salvatore T, Docimo G, Epifani R, Alfano M, Sardu C, Marfella R, Rinaldi L, Sasso FC. Cardiac Hypertrophy: From Pathophysiological Mechanisms to Heart Failure Development. Rev Cardiovasc Med 2022; 23:165. [PMID: 39077592 PMCID: PMC11273913 DOI: 10.31083/j.rcm2305165] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 07/31/2024] Open
Abstract
Cardiac hypertrophy develops in response to increased workload to reduce ventricular wall stress and maintain function and efficiency. Pathological hypertrophy can be adaptive at the beginning. However, if the stimulus persists, it may progress to ventricular chamber dilatation, contractile dysfunction, and heart failure, resulting in poorer outcome and increased social burden. The main pathophysiological mechanisms of pathological hypertrophy are cell death, fibrosis, mitochondrial dysfunction, dysregulation of Ca 2 + -handling proteins, metabolic changes, fetal gene expression reactivation, impaired protein and mitochondrial quality control, altered sarcomere structure, and inadequate angiogenesis. Diabetic cardiomyopathy is a condition in which cardiac pathological hypertrophy mainly develop due to insulin resistance and subsequent hyperglycaemia, associated with altered fatty acid metabolism, altered calcium homeostasis and inflammation. In this review, we summarize the underlying molecular mechanisms of pathological hypertrophy development and progression, which can be applied in the development of future novel therapeutic strategies in both reversal and prevention.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Giovanni Docimo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaella Epifani
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Maria Alfano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| |
Collapse
|
36
|
Sigala DM, Hieronimus B, Medici V, Lee V, Nunez MV, Bremer AA, Cox CL, Price CA, Benyam Y, Abdelhafez Y, McGahan JP, Keim NL, Goran MI, Pacini G, Tura A, Sirlin CB, Chaudhari AJ, Havel PJ, Stanhope KL. The Dose-Response Effects of Consuming High Fructose Corn Syrup-Sweetened Beverages on Hepatic Lipid Content and Insulin Sensitivity in Young Adults. Nutrients 2022; 14:1648. [PMID: 35458210 PMCID: PMC9030734 DOI: 10.3390/nu14081648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 01/27/2023] Open
Abstract
Increased hepatic lipid content and decreased insulin sensitivity have critical roles in the development of cardiometabolic diseases. Therefore, our objective was to investigate the dose-response effects of consuming high fructose corn syrup (HFCS)-sweetened beverages for two weeks on hepatic lipid content and insulin sensitivity in young (18-40 years) adults (BMI 18-35 kg/m2). In a parallel, double-blinded study, participants consumed three beverages/day providing 0% (aspartame: n = 23), 10% (n = 18), 17.5% (n = 16), or 25% (n = 28) daily energy requirements from HFCS. Magnetic resonance imaging for hepatic lipid content and oral glucose tolerance tests (OGTT) were conducted during 3.5-day inpatient visits at baseline and again at the end of a 15-day intervention. During the 12 intervening outpatient days participants consumed their usual diets with their assigned beverages. Significant linear dose-response effects were observed for increases of hepatic lipid content (p = 0.015) and glucose and insulin AUCs during OGTT (both p = 0.0004), and for decreases in the Matsuda (p = 0.0087) and Predicted M (p = 0.0027) indices of insulin sensitivity. These dose-response effects strengthen the mechanistic evidence implicating consumption of HFCS-sweetened beverages as a contributor to the metabolic dysregulation that increases risk for nonalcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- Desiree M. Sigala
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Bettina Hieronimus
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
- Institute for Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany
| | - Valentina Medici
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California—Davis, Sacramento, CA 95817, USA;
| | - Vivien Lee
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Marinelle V. Nunez
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Andrew A. Bremer
- Department of Pediatrics, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA;
| | - Chad L. Cox
- Department of Chemistry, California State University, Sacramento, CA 95819, USA;
- Department of Family and Consumer Sciences, California State University, Sacramento, CA 95819, USA
| | - Candice A. Price
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Yanet Benyam
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Yasser Abdelhafez
- Department of Radiology, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA; (Y.A.); (J.P.M.); (A.J.C.)
| | - John P. McGahan
- Department of Radiology, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA; (Y.A.); (J.P.M.); (A.J.C.)
| | - Nancy L. Keim
- Western Human Nutrition Research Center, United States Department of Agriculture, Davis, CA 95616, USA;
| | - Michael I. Goran
- The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - Giovanni Pacini
- Metabolic Unit, Institute of Neuroscience, National Research Council (CNR), 35127 Padova, Italy;
| | - Andrea Tura
- Liver Imaging Group, Department of Radiology, University of California—San Diego, La Jolla, CA 92093, USA; (A.T.); (C.B.S.)
| | - Claude B. Sirlin
- Liver Imaging Group, Department of Radiology, University of California—San Diego, La Jolla, CA 92093, USA; (A.T.); (C.B.S.)
| | - Abhijit J. Chaudhari
- Department of Radiology, School of Medicine, University of California—Davis, Sacramento, CA 95817, USA; (Y.A.); (J.P.M.); (A.J.C.)
| | - Peter J. Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| | - Kimber L. Stanhope
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California—Davis, Sacramento, CA 95616, USA; (D.M.S.); (B.H.); (V.L.); (M.V.N.); (C.A.P.); (Y.B.); (P.J.H.)
- Department of Nutrition, University of California—Davis, Sacramento, CA 95616, USA
| |
Collapse
|
37
|
Patel SJ, Liu N, Piaker S, Gulko A, Andrade ML, Heyward FD, Sermersheim T, Edinger N, Srinivasan H, Emont MP, Westcott GP, Luther J, Chung RT, Yan S, Kumari M, Thomas R, Deleye Y, Tchernof A, White PJ, Baselli GA, Meroni M, De Jesus DF, Ahmad R, Kulkarni RN, Valenti L, Tsai L, Rosen ED. Hepatic IRF3 fuels dysglycemia in obesity through direct regulation of Ppp2r1b. Sci Transl Med 2022; 14:eabh3831. [PMID: 35320000 PMCID: PMC9162056 DOI: 10.1126/scitranslmed.abh3831] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation has profound but poorly understood effects on metabolism, especially in the context of obesity and nonalcoholic fatty liver disease (NAFLD). Here, we report that hepatic interferon regulatory factor 3 (IRF3) is a direct transcriptional regulator of glucose homeostasis through induction of Ppp2r1b, a component of serine/threonine phosphatase PP2A, and subsequent suppression of glucose production. Global ablation of IRF3 in mice on a high-fat diet protected against both steatosis and dysglycemia, whereas hepatocyte-specific loss of IRF3 affects only dysglycemia. Integration of the IRF3-dependent transcriptome and cistrome in mouse hepatocytes identifies Ppp2r1b as a direct IRF3 target responsible for mediating its metabolic actions on glucose homeostasis. IRF3-mediated induction of Ppp2r1b amplified PP2A activity, with subsequent dephosphorylation of AMPKα and AKT. Furthermore, suppression of hepatic Irf3 expression with antisense oligonucleotides reversed obesity-induced insulin resistance and restored glucose homeostasis in obese mice. Obese humans with NAFLD displayed enhanced activation of liver IRF3, with reversion after bariatric surgery. Hepatic PPP2R1B expression correlated with HgbA1C and was elevated in obese humans with impaired fasting glucose. We therefore identify the hepatic IRF3-PPP2R1B axis as a causal link between obesity-induced inflammation and dysglycemia and suggest an approach for limiting the metabolic dysfunction accompanying obesity-associated NAFLD.
Collapse
Affiliation(s)
- Suraj J. Patel
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Division of Digestive and Liver Diseases, Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nan Liu
- Harvard Medical School, Boston, MA 02115, USA
- Cancer and Blood Disorders Center, Dana Farber Cancer Institute and Boston Children’s Hospital, Boston, MA 02215, USA
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
| | - Sam Piaker
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Anton Gulko
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Maynara L. Andrade
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Frankie D. Heyward
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Tyler Sermersheim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Nufar Edinger
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Harini Srinivasan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Margo P. Emont
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Gregory P. Westcott
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jay Luther
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Raymond T. Chung
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shuai Yan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Manju Kumari
- Department of Cardiology, Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Reeby Thomas
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Yann Deleye
- Duke Molecular Physiology Institute and Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - André Tchernof
- Institut Universitaire de Cardiologie and Pneumologie de Québec–Université Laval (IUCPQUL), Québec City, Canada
| | - Phillip J. White
- Duke Molecular Physiology Institute and Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido A. Baselli
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario F. De Jesus
- Harvard Medical School, Boston, MA 02115, USA
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rohit N. Kulkarni
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Universita degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linus Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Evan D. Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
38
|
Mahmoudi A, Butler AE, Majeed M, Banach M, Sahebkar A. Investigation of the Effect of Curcumin on Protein Targets in NAFLD Using Bioinformatic Analysis. Nutrients 2022; 14:1331. [PMID: 35405942 PMCID: PMC9002953 DOI: 10.3390/nu14071331] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a prevalent metabolic disorder. Defects in function/expression of genes/proteins are critical in initiation/progression of NAFLD. Natural products may modulate these genes/proteins. Curcumin improves steatosis, inflammation, and fibrosis progression. Here, bioinformatic tools, gene−drug and gene-disease databases were utilized to explore targets, interactions, and pathways through which curcumin could impact NAFLD. METHODS: Significant curcumin−protein interaction was identified (high-confidence:0.7) in the STITCH database. Identified proteins were investigated to determine association with NAFLD. gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were analyzed for significantly involved targets (p < 0.01). Specificity of obtained targets with NAFLD was estimated and investigated in Tissue/Cells−gene associations (PanglaoDB Augmented 2021, Mouse Gene Atlas) and Disease−gene association-based EnrichR algorithms (Jensen DISEASES, DisGeNET). RESULTS: Two collections were constructed: 227 protein−curcumin interactions and 95 NAFLD-associated genes. By Venn diagram, 14 significant targets were identified, and their biological pathways evaluated. Based on gene ontology, most targets involved stress and lipid metabolism. KEGG revealed chemical carcinogenesis, the AGE-RAGE signaling pathway in diabetic complications and NAFLD as the most common significant pathways. Specificity to diseases database (EnrichR algorithm) revealed specificity for steatosis/steatohepatitis. CONCLUSION: Curcumin may improve, or inhibit, progression of NAFLD through activation/inhibition of NAFLD-related genes.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran;
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain;
| | | | - Maciej Banach
- Nephrology and Hypertension, Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 93-338 Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
| |
Collapse
|
39
|
Deletion of RNF186 expression suppresses diet-induced hepatic steatosis by regulating insulin activity. iScience 2022; 25:103859. [PMID: 35198905 PMCID: PMC8850801 DOI: 10.1016/j.isci.2022.103859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 11/20/2022] Open
Abstract
RING finger protein186 (RNF186) is dramatically upregulated in steatotic livers. The physiological role of RNF186 in non-alcoholic fatty liver disease (NAFLD) remains obscure. Here, we found that hepatocyte-specific RNF186 knockout (RNF186LKO) mice were protected from HFD-induced obesity. RNF186 ablation in liver suppressed inflammatory responses and ER stress and alleviated insulin tolerance, leading to improved glucose and lipid metabolism under HFD conditions. RNA-seq and western blot analyses revealed a significant downregulation of peroxisome proliferator-activated receptor γ, stearoyl-CoA desaturase 1, and cluster of differentiation 36 in the liver of RNF186 knockout mice consuming HFD. RNF186 deletion in liver results in less weight gain during HFD feeding and is associated with reduced liver fat, inflammation, and improved glucose and insulin tolerance. In contrast, upregulation of RNF186 in C57BL/6J mice livers impaired lipid metabolism and insulin tolerance. The collective results suggest that RNF186 may be a potential regulator of NAFLD in obesity. RNF186 deficiency on high-fat diet alleviates liver steatosis and insulin tolerance RNF186 increased hepatic TG accumulation and impaired insulin sensitivity RNF186 ablation suppresses hepatic inflammation associated with high-fat diet RNF186 maybe a potential regulator of NAFLD in obesity
Collapse
|
40
|
Mansour R, El-Fayoumi HM, Fahmy A, Ibrahim IAAEH. Oleic acid acutely impairs glucose homeostasis in standard chow diet but not high-fructose, high-fat diet-fed mice by acting on free fatty acid receptor 1. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20710s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
41
|
Liu X, Lin J, Zhang H, Khan NU, Zhang J, Tang X, Cao X, Shen L. Oxidative Stress in Autism Spectrum Disorder-Current Progress of Mechanisms and Biomarkers. Front Psychiatry 2022; 13:813304. [PMID: 35299821 PMCID: PMC8921264 DOI: 10.3389/fpsyt.2022.813304] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorder (ASD) is a type of neurodevelopmental disorder that has been diagnosed in an increasing number of children around the world. Existing data suggest that early diagnosis and intervention can improve ASD outcomes. However, the causes of ASD remain complex and unclear, and there are currently no clinical biomarkers for autism spectrum disorder. More mechanisms and biomarkers of autism have been found with the development of advanced technology such as mass spectrometry. Many recent studies have found a link between ASD and elevated oxidative stress, which may play a role in its development. ASD is caused by oxidative stress in several ways, including protein post-translational changes (e.g., carbonylation), abnormal metabolism (e.g., lipid peroxidation), and toxic buildup [e.g., reactive oxygen species (ROS)]. To detect elevated oxidative stress in ASD, various biomarkers have been developed and employed. This article summarizes recent studies about the mechanisms and biomarkers of oxidative stress. Potential biomarkers identified in this study could be used for early diagnosis and evaluation of ASD intervention, as well as to inform and target ASD pharmacological or nutritional treatment interventions.
Collapse
Affiliation(s)
- Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Jun Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen, China
| |
Collapse
|
42
|
Singh SP, Anirvan P, Khandelwal R, Satapathy SK. Nonalcoholic Fatty Liver Disease (NAFLD) Name Change: Requiem or Reveille? J Clin Transl Hepatol 2021; 9:931-938. [PMID: 34966656 PMCID: PMC8666378 DOI: 10.14218/jcth.2021.00174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/21/2021] [Accepted: 07/18/2021] [Indexed: 12/04/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects about a quarter of the world's population and poses a major health and economic burden globally. Recently, there have been hasty attempts to rename NAFLD to metabolic-associated fatty liver disease (MAFLD) despite the fact that there is no scientific rationale for this. Quest for a "positive criterion" to diagnose the disease and destigmatizing the disease have been the main reasons put forth for the name change. A close scrutiny of the pathogenesis of NAFLD would make it clear that NAFLD is a heterogeneous disorder, involving different pathogenic mechanisms of which metabolic dysfunction-driven hepatic steatosis is only one. Replacing NAFLD with MAFLD would neither enhance the legitimacy of clinical practice and clinical trials, nor improve clinical care or move NAFLD research forward. Rather than changing the nomenclature without a strong scientific backing to support such a change, efforts should be directed at understanding NAFLD pathogenesis across diverse populations and ethnicities which could potentially help develop newer therapeutic options.
Collapse
Affiliation(s)
- Shivaram P. Singh
- Department of Gastroenterology, Sriram Chandra Bhanj Medical College, Cuttack, Odisha, India
| | - Prajna Anirvan
- Department of Gastroenterology, Sriram Chandra Bhanj Medical College, Cuttack, Odisha, India
| | - Reshu Khandelwal
- Department of Gastroenterology, Sriram Chandra Bhanj Medical College, Cuttack, Odisha, India
| | - Sanjaya K. Satapathy
- Division of Hepatology, Sandra Atlas Bass Center for Liver Diseases and Transplantation, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Manhasset, NY, USA
| |
Collapse
|
43
|
Saed CT, Greenwell AA, Tabatabaei Dakhili SA, Gopal K, Eaton F, Ussher JR. The antianginal ranolazine does not confer beneficial actions against hepatic steatosis in male mice subjected to high-fat diet and streptozotocin induced type 2 diabetes. Can J Physiol Pharmacol 2021; 100:393-401. [PMID: 34851748 DOI: 10.1139/cjpp-2021-0559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by the accumulation of excess fat in the liver in the absence of alcohol and increases one's risk for both diabetes and cardiovascular disease (e.g. angina). We have shown that the second-line anti-anginal therapy, ranolazine, mitigates obesity-induced NAFLD, and our aim was to determine whether these actions of ranolazine also extend to NAFLD associated with type 2 diabetes (T2D). 8-week-old male C57BL/6J mice were fed either a low-fat diet or a high-fat diet for 15-weeks, with a single dose of streptozotocin (STZ; 75 mg/kg) administered in the high-fat diet fed mice at 4-weeks to induce experimental T2D. Mice were treated with either vehicle control or ranolazine during the final 7-weeks (50 mg/kg once daily). We assessed glycemia via monitoring glucose tolerance, insulin tolerance, and pyruvate tolerance, whereas hepatic steatosis was assessed via quantifying triacylglycerol content. We observed that ranolazine did not improve glycemia in mice with experimental T2D, while also having no impact on hepatic triacylglycerol content. Therefore, the salutary actions of ranolazine against NAFLD may be limited to obese individuals but not those who are obese with T2D.
Collapse
Affiliation(s)
| | | | | | - Keshav Gopal
- University of Alberta, 3158, University of Alberta, Edmonton, Alberta, Canada, T6G2E1;
| | - Farah Eaton
- University of Alberta, 3158, Pharmacy, 2-055 Katz, Edmonton, Alberta, Canada, T6G 2R3.,Canada;
| | - John R Ussher
- University of Alberta, 2-020C Katz Centre for Pharmacy and Health Research, Edmonton, Alberta, Canada, T6G 2E1;
| |
Collapse
|
44
|
Hypothyroidism-Associated Dyslipidemia: Potential Molecular Mechanisms Leading to NAFLD. Int J Mol Sci 2021; 22:ijms222312797. [PMID: 34884625 PMCID: PMC8657790 DOI: 10.3390/ijms222312797] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/31/2022] Open
Abstract
Thyroid hormones control lipid metabolism by exhibiting specific effects on the liver and adipose tissue in a coordinated manner. Different diseases of the thyroid gland can result in hypothyroidism. Hypothyroidism is frequently associated with dyslipidemia. Hypothyroidism-associated dyslipidemia subsequently results in intrahepatic accumulation of fat, leading to nonalcoholic fatty liver disease (NAFLD), which leads to the development of hepatic insulin resistance. The prevalence of NAFLD in the western world is increasing, and evidence of its association with hypothyroidism is accumulating. Since hypothyroidism has been identified as a modifiable risk factor of NAFLD and recent data provides evidence that selective thyroid hormone receptor β (THR-β) agonists are effective in the treatment of dyslipidemia and NAFLD, interest in potential therapeutic options for NAFLD targeting these receptors is growing. In this review, we summarize current knowledge regarding clinical and molecular data exploring the association of hypothyroidism, dyslipidemia and NAFLD.
Collapse
|
45
|
Mohamed RMSM, Elshazly SM, Nafea OE, Abd El Motteleb DM. Comparative cardioprotective effects of carvedilol versus atenolol in a rat model of cardiorenal syndrome type 4. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:2117-2128. [PMID: 34398250 DOI: 10.1007/s00210-021-02130-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022]
Abstract
The incidence of chronic kidney disease is escalating; cardiorenal syndrome (CRS) type 4 is gaining a major health concern causing significant morbidity and mortality, putting major burdens on the healthcare system. This study was designed to compare the cardioprotective effects of carvedilol versus atenolol against CRS type 4 induced by subtotal 5/6 nephrectomy in rats and to explore the underlying mechanisms. Immediately after surgery, carvedilol (20 mg/kg/day) or atenolol (20 mg/kg/day) was added to drinking water for 10 weeks. Carvedilol was more effective than atenolol in improving kidney functions, decreasing elevated blood pressures, attenuating cardiac hypertrophy, reducing serum brain natriuretic peptide, and diminished cardiac fibrous tissue deposition. However, carvedilol was equivalent to atenolol in modulating β1-adrenergic receptors (β1ARs) and cardiac diacylglycerol (DAG) signaling, but carvedilol was superior in modulating β-arrestin2, phosphatidyl inositol 4,5 bisphosphates (PIP2), and caspase 3 levels. Carvedilol has superior cardioprotective effects than atenolol in a rat model of CRS type 4. These protective effects are mediated through modulating cardiac β1ARs/β-arrestin2/PIP2/DAG as well as abating cardiac apoptotic signaling pathways (caspase3/pS473 protein kinase B (Akt)).
Collapse
Affiliation(s)
- Rasha M S M Mohamed
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Shimaa M Elshazly
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ola E Nafea
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia.
| | | |
Collapse
|
46
|
Quantitative Proteomics and Phosphoproteomics Reveal TNF-α-Mediated Protein Functions in Hepatocytes. Molecules 2021; 26:molecules26185472. [PMID: 34576943 PMCID: PMC8464716 DOI: 10.3390/molecules26185472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 11/18/2022] Open
Abstract
Increased secretion of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNFα), is often associated with adipose tissue dysregulation, which often accompanies obesity. High levels of TNFα have been linked to the development of insulin resistance in several tissues and organs, including skeletal muscle and the liver. In this study, we examined the complex regulatory roles of TNFα in murine hepatocytes utilizing a combination of global proteomic and phosphoproteomic analyses. Our results show that TNFα promotes extensive changes not only of protein levels, but also the dynamics of their downstream phosphorylation signaling. We provide evidence that TNFα induces DNA replication and promotes G1/S transition through activation of the MAPK pathway. Our data also highlight several other novel proteins, many of which are regulated by phosphorylation and play a role in the progression and development of insulin resistance in hepatocytes.
Collapse
|
47
|
Korac B, Kalezic A, Pekovic-Vaughan V, Korac A, Jankovic A. Redox changes in obesity, metabolic syndrome, and diabetes. Redox Biol 2021; 42:101887. [PMID: 33579666 PMCID: PMC8113039 DOI: 10.1016/j.redox.2021.101887] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
"Life is an instantaneous encounter of circulating matter and flowing energy" (Jean Giaja, Serbian physiologist), is one of the most elegant definitions not only of life but the relationship of redox biology and metabolism. Their evolutionary liaison has created inseparable yet dynamic homeostasis in health, which, when disrupted, leads to disease. This interconnection is even more pertinent today, in an era of increasing metabolic diseases of epidemic proportions such as obesity, metabolic syndrome, and diabetes. Despite great advances in understanding the molecular mechanisms of redox and metabolic regulation, we face significant challenges in preventing, diagnosing, and treating metabolic diseases. The etiological association and temporal overlap of these syndromes present significant challenges for the discrimination of appropriate clinical biomarkers for diagnosis, treatment, and outcome prediction. These multifactorial, multiorgan metabolic syndromes with complex etiopathogenic mechanisms are accompanied by disturbed redox equilibrium in target tissues and circulation. Free radicals and reactive species are considered both a causal factor and a consequence of disease status. Thus, determining the subtypes and levels of free radicals and reactive species, oxidatively damaged biomolecules (lipids, proteins, and nucleic acids) and antioxidant defense components as well as redox-sensitive transcription factors and fluxes of redox-dependent metabolic pathways will help define existing and establish novel redox biomarkers for stratifying metabolic diseases. This review aims to discuss diverse redox/metabolic aspects in obesity, metabolic syndrome, and diabetes, with the imperative to help establish a platform for emerging and future redox-metabolic biomarkers research in precision medicine. Future research warrants detailed investigations into the status of redox biomarkers in healthy subjects and patients, including the use of emerging 'omic' profiling technologies (e.g., redox proteomes, lipidomes, metabolomes, and transcriptomes), taking into account the influence of lifestyle (diet, physical activity, sleep, work patterns) as well as circadian ~24h fluctuations in circulatory factors and metabolites.
Collapse
Affiliation(s)
- Bato Korac
- Department of Physiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia; Center for Electron Microscopy, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia.
| | - Andjelika Kalezic
- Department of Physiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia
| | - Vanja Pekovic-Vaughan
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, William Henry Duncan Building, University of Liverpool, L7 8TX, Liverpool, UK
| | - Aleksandra Korac
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia.
| |
Collapse
|
48
|
Dallio M, Romeo M, Gravina AG, Masarone M, Larussa T, Abenavoli L, Persico M, Loguercio C, Federico A. Nutrigenomics and Nutrigenetics in Metabolic- (Dysfunction) Associated Fatty Liver Disease: Novel Insights and Future Perspectives. Nutrients 2021; 13:1679. [PMID: 34063372 PMCID: PMC8156164 DOI: 10.3390/nu13051679] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic- (dysfunction) associated fatty liver disease (MAFLD) represents the predominant hepatopathy and one of the most important systemic, metabolic-related disorders all over the world associated with severe medical and socio-economic repercussions due to its growing prevalence, clinical course (steatohepatitis and/or hepatocellular-carcinoma), and related extra-hepatic comorbidities. To date, no specific medications for the treatment of this condition exist, and the most valid recommendation for patients remains lifestyle change. MAFLD has been associated with metabolic syndrome; its development and progression are widely influenced by the interplay between genetic, environmental, and nutritional factors. Nutrigenetics and nutrigenomics findings suggest nutrition's capability, by acting on the individual genetic background and modifying the specific epigenetic expression as well, to influence patients' clinical outcome. Besides, immunity response is emerging as pivotal in this multifactorial scenario, suggesting the interaction between diet, genetics, and immunity as another tangled network that needs to be explored. The present review describes the genetic background contribution to MAFLD onset and worsening, its possibility to be influenced by nutritional habits, and the interplay between nutrients and immunity as one of the most promising research fields of the future in this context.
Collapse
Affiliation(s)
- Marcello Dallio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Mario Romeo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Antonietta Gerarda Gravina
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Mario Masarone
- Department of Medicine and Surgery, University of Salerno, Via Allende, 84081 Baronissi, Italy; (M.M.); (M.P.)
| | - Tiziana Larussa
- Department of Health Sciences, University Magna Graecia, viale Europa, 88100 Catanzaro, Italy; (T.L.); (L.A.)
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia, viale Europa, 88100 Catanzaro, Italy; (T.L.); (L.A.)
| | - Marcello Persico
- Department of Medicine and Surgery, University of Salerno, Via Allende, 84081 Baronissi, Italy; (M.M.); (M.P.)
| | - Carmelina Loguercio
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| | - Alessandro Federico
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via S. Pansini 5, 80131 Naples, Italy; (M.R.); (A.G.G.); (C.L.); (A.F.)
| |
Collapse
|
49
|
Wigger D, Schumacher F, Schneider-Schaulies S, Kleuser B. Sphingosine 1-phosphate metabolism and insulin signaling. Cell Signal 2021; 82:109959. [PMID: 33631318 DOI: 10.1016/j.cellsig.2021.109959] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022]
Abstract
Insulin is the main anabolic hormone secreted by β-cells of the pancreas stimulating the assimilation and storage of glucose in muscle and fat cells. It modulates the postprandial balance of carbohydrates, lipids and proteins via enhancing lipogenesis, glycogen and protein synthesis and suppressing glucose generation and its release from the liver. Resistance to insulin is a severe metabolic disorder related to a diminished response of peripheral tissues to the insulin action and signaling. This leads to a disturbed glucose homeostasis that precedes the onset of type 2 diabetes (T2D), a disease reaching epidemic proportions. A large number of studies reported an association between elevated circulating fatty acids and the development of insulin resistance. The increased fatty acid lipid flux results in the accumulation of lipid droplets in a variety of tissues. However, lipid intermediates such as diacylglycerols and ceramides are also formed in response to elevated fatty acid levels. These bioactive lipids have been associated with the pathogenesis of insulin resistance. More recently, sphingosine 1-phosphate (S1P), another bioactive sphingolipid derivative, has also been shown to increase in T2D and obesity. Although many studies propose a protective role of S1P metabolism on insulin signaling in peripheral tissues, other studies suggest a causal role of S1P on insulin resistance. In this review, we critically summarize the current state of knowledge of S1P metabolism and its modulating role on insulin resistance. A particular emphasis is placed on S1P and insulin signaling in hepatocytes, skeletal muscle cells, adipocytes and pancreatic β-cells. In particular, modulation of receptors and enzymes that regulate S1P metabolism can be considered as a new therapeutic option for the treatment of insulin resistance and T2D.
Collapse
Affiliation(s)
- Dominik Wigger
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany
| | | | - Burkhard Kleuser
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany; Institute of Nutritional Science, Nutritional Toxicology, University of Potsdam, Nuthetal, Germany.
| |
Collapse
|
50
|
Storck LJ, Meffert PJ, Rausch J, Gärtner S, Aghdassi AA, Kühn JP, Kraft M, Pietzner M, Lerch MM, Steveling A. Efficiency of a 15-Week Weight-Loss Program, Including a Low-Calorie Formula Diet, on Glycemic Control in Patients with Type 2 Diabetes Mellitus and Overweight or Obesity. Obes Facts 2021; 14:1-11. [PMID: 33601371 PMCID: PMC7983589 DOI: 10.1159/000511453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Patients who are overweight or obese have an increased risk of developing type 2 diabetes mellitus (T2DM). Weight loss can have a positive effect on glycemic control. OBJECTIVE We aimed to investigate glycemic control in patients with T2DM and overweight or obesity during a structured weight-loss program. METHODS This was a prospective, interventional study. We recruited 36 patients (14 men and 22 women) with a median age of 58.5 years and median body mass index (BMI) of 34.1, to a 15-week structured weight-loss program with a low-calorie (800 kcal) formula diet for 6 weeks. The primary end point, HbA1c level, and secondary end points, anthropometric data, medication, and safety, were assessed weekly. Laboratory values and quality of life were assessed at baseline and after 15 weeks. RESULTS HbA1c decreased from 7.3% at baseline to 6.5% at 15 weeks (p < 0.001), median body weight by 11.9 kg (p < 0.001), median BMI by 4.3 (p < 0.001) and median waist circumference by 11.0 cm (p < 0.001). Two participants discontinued insulin therapy, 4 could reduce their dosage of oral antidiabetic agents, and 6 completely discontinued their antidiabetic medication. Insulin dose decreased from 0.63 (0.38-0.89) to 0.39 (0.15-0.70) units/kg body weight (p < 0.001). No patient experienced hypoglycemic episodes or hospital emergency visits. Triglycerides and total cholesterol decreased as well as surrogate markers of liver function. However, the levels of high-density and low-density lipoprotein cholesterol (HDL-C and LDL-C) as well as uric acid remain unchanged. Regarding quality of life, the median physical health score increased from 44.5 (39.7-51.4) at baseline to 48.0 (43.1-55.3; p = 0.007), and the median mental health score decreased from 42.1 (36.1-46.7) to 37.4 (30.3-43.7; p = 0.004). CONCLUSIONS A structured weight-loss program is effective in the short term in reducing HbA1c, weight, and antidiabetic medication in patients with T2DM who are overweight or obese. Levels of HDL-C and LDL-C were not affected by short-term weight loss. The decline in mental health and the long-term effects of improved glycemic control require further trials.
Collapse
Affiliation(s)
- Lena J Storck
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
- Department of Medicine, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Peter J Meffert
- Institute of Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Corvus, Statistical Consulting, Altkalen, Germany
| | - Janine Rausch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Simone Gärtner
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Ali A Aghdassi
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Jens-Peter Kühn
- Institute of Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
- Institute of Radiology, Carl-Gustav Carus University Dresden, Dresden, Germany
| | - Matthias Kraft
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
- Medizinische Klinik I, Vinzentius-Krankenhaus Landau, Landau, Germany
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany,
| | - Antje Steveling
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|