1
|
Boulos M, Mousa RS, Jeries N, Simaan E, Alam K, Bulus B, Assy N. Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. Int J Mol Sci 2025; 26:3448. [PMID: 40244398 PMCID: PMC11989262 DOI: 10.3390/ijms26073448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic syndrome (MetS) and metabolic dysfunction-associated steatotic liver disease (MASLD) are closely related, with rapidly increasing prevalence globally, driving significant public health concerns. Both conditions share common pathophysiological mechanisms such as insulin resistance (IR), adipose tissue dysfunction, oxidative stress, and gut microbiota dysbiosis, which contribute to their co-occurrence and progression. While the clinical implications of this overlap, including increased cardiovascular, renal, and hepatic risk, are well recognized, current diagnostic and therapeutic approaches remain insufficient due to the clinical and individuals' heterogeneity and complexity of these diseases. This review aims to provide an in-depth exploration of the molecular mechanisms linking MetS and MASLD, identify critical gaps in our understanding, and highlight existing challenges in early detection and treatment. Despite advancements in biomarkers and therapeutic interventions, the need for a comprehensive, integrated approach remains. The review also discusses emerging therapies targeting specific pathways, the potential of precision medicine, and the growing role of artificial intelligence in enhancing research and clinical management. Future research is urgently needed to combine multi-omics data, precision medicine, and novel biomarkers to better understand the complex interactions between MetS and MASLD. Collaborative, multidisciplinary efforts are essential to develop more effective diagnostic tools and therapies to address these diseases on a global scale.
Collapse
Affiliation(s)
- Mariana Boulos
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Rabia S. Mousa
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Nizar Jeries
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Elias Simaan
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Klode Alam
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Bulus Bulus
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
| | - Nimer Assy
- Internal Medicine Department, Galilee Medical Centre, Nahariya 221001, Israel; (R.S.M.); (N.J.); (E.S.); (K.A.); (B.B.); (N.A.)
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
2
|
Qiu X, Wu W, Zhang S, Huang C, Lin D. 3-Hydroxybutyrate Promotes Myoblast Proliferation and Differentiation through Energy Metabolism and GPR109a-Mediated Ca 2+-NFAT Signaling Pathways. J Proteome Res 2025; 24:2063-2080. [PMID: 40099866 DOI: 10.1021/acs.jproteome.4c01150] [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] [Indexed: 03/20/2025]
Abstract
Skeletal muscle wasting is a critical clinical problem associated with several diseases that significantly impair patient outcomes due to the progressive loss of muscle mass and function. This study explores the potential of 3-hydroxybutyrate (3-HB) as a therapeutic agent to counteract muscle atrophy by promoting the proliferation and differentiation of C2C12 myoblasts. Using nuclear magnetic resonance (NMR)-based metabolomics analysis, we uncover the underlying mechanisms by which 3-HB exerts its effects. Our findings demonstrate that 3-HB exerts its effects through two distinct mechanisms: as a metabolic substrate and as a signaling molecule. As a metabolic substrate, 3-HB enhances myoblast energy efficiency by stimulating the expression of G protein-coupled receptor 109a (GPR109a), which subsequently upregulates the 3-HB transporters MCT1 and CD147, the utilization enzyme OXCT1, and phosphorylated AMPK, thereby increasing ATP production. As a signaling molecule, 3-HB activates GPR109a, promoting calcium influx, improving calcium homeostasis, and increasing the expression of Ca2+-related proteins such as CAMKK2. This signaling cascade activates calcineurin (CaN), facilitating NFAT translocation to the nucleus and gene expression that drives myoblast proliferation and differentiation. By elucidating the dual regulatory roles of 3-HB in energy metabolism and cellular signaling, this study not only advances our understanding of muscle physiology but also highlights the potential of 3-HB as a novel therapeutic approach for the prevention or treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Xu Qiu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Wenfang Wu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuya Zhang
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
3
|
Duarte GBS, Pascoal GDFL, Rogero MM. Polymorphisms Involved in Insulin Resistance and Metabolic Inflammation: Influence of Nutrients and Dietary Interventions. Metabolites 2025; 15:245. [PMID: 40278374 PMCID: PMC12029114 DOI: 10.3390/metabo15040245] [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: 02/28/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Insulin resistance (IR) is a metabolic disorder characterized by an impaired response to insulin. This condition is associated with excess adiposity and metabolic inflammation, contributing to an increased risk for related chronic diseases. Single-nucleotide polymorphisms (SNPs) can affect genes related to metabolic pathways which are related to IR and the individual response to nutrients and dietary patterns, affecting metabolic inflammation and insulin sensitivity. This narrative review explores the current evidence on interactions between genetic variants and dietary factors, specifically their effects in modulating IR and metabolic inflammation. A comprehensive search of the literature was conducted in PubMed, Google Scholar, and Web of Science, and a total of 95 articles were reviewed. The key findings reveal that SNPs in the TCF7L2, ADIPOQ, and TNF genes significantly influence metabolic responses and modulate the effects of the Mediterranean diet on biomarkers of inflammation and IR. Genotype-dependent variations in IR and inflammation biomarkers were observed in the response to different diets for SNPs in the TCF7L2, ADIPOQ, and TNF genes. Additionally, polygenic risk scores (PRSs) can also predict the response to the intake of nutrients and specific diets, and offer a promising tool for assessing genetic predisposition to IR. This review underscores the pivotal role of an individual's genetic background in the effects of their nutrient intake and in the responses to dietetic interventions, thereby laying the foundation for personalized and effective nutritional strategies tailored to each individual's necessity in mitigating IR and its associated risk factors for chronic diseases.
Collapse
Affiliation(s)
| | | | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (G.B.S.D.); (G.d.F.L.P.)
| |
Collapse
|
4
|
Montoya Castillo M, Martínez Quiroz WDJ, Suarez-Ortegón MF, Higuita-Gutiérrez LF. Waist-to-Height Ratio, Waist Circumference, and Body Mass Index in Relation to Full Cardiometabolic Risk in an Adult Population from Medellin, Colombia. J Clin Med 2025; 14:2411. [PMID: 40217861 PMCID: PMC11989366 DOI: 10.3390/jcm14072411] [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: 02/28/2025] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Few studies have compared the associations of different adiposity markers with cardiometabolic risk factors in individuals without diabetes or cardiovascular disease (CVD), particularly in South America. Moreover, the associations with more severe cardiometabolic risk, defined by the simultaneous presence of altered glycemia, blood pressure, and dyslipidemia, remain unknown. We examined whether the waist-to-height ratio (W-HtR), waist circumference (WC), and BMI were independently associated with cardiometabolic risk in a chronic disease prevention program in Medellín, Colombia. Methods: A cross-sectional study was conducted in 29,236 adults (age: 19-121 years) without diabetes or CVD. Exposures included increased W-HtR (>0.5), increased WC (≥80 cm for women, ≥90 cm for men), and overweight/obesity. The outcomes were dyslipidemia, elevated glycemia, high blood pressure, and full cardiometabolic risk (FCMR), defined as the presence of all three factors. Logistic regressions adjusted for sociodemographic and lifestyle covariates and additional adiposity markers were used. Cubic spline analyses examined the shape of associations. Results: Most individuals were over 40 years old (97.6%), only 40 were ≥100 years, and 16.5% (n = 4821) had FCMR. Increased W-HtR tripled the odds of FCMR compared with normal W-HtR (OR: 3.04, 95%CI: 2.45-3.77, p < 0.001). Increased WC doubled the odds of FCMR (p < 0.001). W-HtR remained the strongest predictor after adjusting for WC (OR: 1.99, 95%CI: 1.59-2.50) and BMI (OR: 2.48, 95%CI: 1.99-3.08). Cubic spline analyses showed a linear association between W-HtR and FCMR, whereas the BMI-FCMR association plateaued at approximately 30 kg/m2. Conclusions: In this cross-sectional study of a large middle-to-older-aged cohort, W-HtR was the strongest adiposity marker correlated with cardiometabolic risk.
Collapse
Affiliation(s)
| | | | - Milton Fabian Suarez-Ortegón
- Departamento de Alimentación y Nutrición, Facultad de Ciencias de la Salud, Pontificia Universidad Javeriana Seccional Cali, Cali 760031, Colombia
| | | |
Collapse
|
5
|
Xu M, Lv D, Wei H, Li Z, Jin S, Liu Q, Zhang Y, Liu Y. Effects of antidiabetic agents on lipid metabolism of skeletal muscle: A narrative review. Diabetes Obes Metab 2025; 27:1693-1707. [PMID: 39807619 DOI: 10.1111/dom.16189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
Metabolic syndrome-related diseases frequently involve disturbances in skeletal muscle lipid metabolism. The accumulation of lipid metabolites, lipid-induced mitochondrial stress in skeletal muscle cells, as well as the inflammation of adjacent adipose tissue, are associated with the development of insulin resistance and metabolic dysfunction. Consequently, when antidiabetic medications are used to treat various chronic conditions related to hyperglycaemia, the impact on skeletal muscle lipid metabolism should not be overlooked. However, current research has predominantly focused on muscle mass rather than skeletal muscle lipid metabolism and its interplay with glucose metabolism. In this review, we summarised the latest research on the effects of antidiabetic drugs and certain natural compounds with antidiabetic activity on skeletal muscle lipid metabolism, focusing on data from preclinical to clinical studies. Given the widespread use of antidiabetic drugs, a better understanding of their effects on skeletal muscle lipid metabolism merits further attention in future research.
Collapse
Affiliation(s)
- Ming Xu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Dongqing Lv
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hongxia Wei
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhe Li
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shuqing Jin
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Qinhao Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
6
|
Peng H, Wang B, Yang W, Jia R, Luo Y, Chen W. Association Between Triglyceride and Incident Diabetes Mellitus: A Secondary Retrospective Analysis Based on a Chinese Cohort Study. J Multidiscip Healthc 2025; 18:1779-1790. [PMID: 40171237 PMCID: PMC11960458 DOI: 10.2147/jmdh.s510549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 04/03/2025] Open
Abstract
Background The association between triglyceride(TG) levels and the risk of diabetes mellitus (DM) continues to be a subject of considerable interest and debate within the scientific community. To date, there has been a lack of studies specifically examining this relationship within the Chinese population. This study seeks to elucidate the correlation between TG levels and the incidence of DM among the Chinese demographic. Methods This study constitutes a secondary analysis of a retrospective cohort investigation comprising 202,888 Chinese participants who were free of DM at baseline and were subsequently followed from 2010 to 2016. Cox regression method and sensitivity analyses were used to examine the relationship between TG levels and DM. To examine the potential non-linear relationship between TG levels and the incidence of DM, Cox proportional hazards regression incorporating cubic spline functions and smooth curve fitting was employed. Additionally, a two-piece Cox proportional hazards regression model was utilized to identify the inflection point at which TG levels influence the risk of developing DM. Results In participants with DM, baseline TG levels were elevated. After adjusting for confounding variables, baseline TG levels were positively associated with incident DM. (HR:1.25,95% CI:1.21-1.30,P<0.001). In addition, we conducted sensitivity analyses to ensure the results were robust. There was a 88% increase in DM risk from the top TG tertile to the bottom TG tertile.Our research discovered a significant link between TG and DM when TG levels were below 1.27 mmol/L (HR:2.35, 95% CI: 1.95-2.83,P < 0.001). Conclusion This study shows that TG was positively and non-linearly associated with the risk of DM after adjusting for other confounding factors.Below 1.27 mmol/L, increasing TG levels greatly heighten the risk of DM, whereas above this level, the risk is lower.
Collapse
Affiliation(s)
- Hui Peng
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Bin Wang
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Wei Yang
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Rui Jia
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Youlian Luo
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Weifeng Chen
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| |
Collapse
|
7
|
Lee YG, Kim D. Tomatine Improves Glucose Metabolism and Mitochondrial Respiration in Insulin-Resistant Hepatocyte Cell Lines AML12 and HepG2 via an AMP-Activated Protein Kinase-Dependent Pathway. Cells 2025; 14:329. [PMID: 40072058 PMCID: PMC11898437 DOI: 10.3390/cells14050329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/15/2025] Open
Abstract
Insulin resistance (IR) disrupts hepatic glucose metabolism and mitochondrial function, which contributes to metabolic disorders. The present study examined the effects of tomatine on glucose metabolism in high-glucose-induced IR hepatocytes and explored its underlying mechanisms using AML12 and HepG2 cell models. The results showed that tomatine did not exhibit cytotoxic effects. Under IR conditions, tomatine dose-dependently improved glucose metabolism by enhancing glucose consumption and restoring the mRNA expression of the glucose transporter Glut2 and gluconeogenesis-related genes (Pepck and G6pase). Mechanistically, tomatine activated the phosphorylation of AMP-activated protein kinase (AMPK) and upregulated the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), reversing the IR-induced suppression of the AMPK/PGC1α pathway. In addition, tomatine enhanced mitochondrial oxidative function by restoring the oxygen consumption rate, increasing ATP production, and upregulating mitochondrial oxidative phosphorylation complex proteins. Both genetic and pharmacological inhibition of AMPK abolished these beneficial effects, confirming its central role in mediating tomatine's actions. Overall, our findings suggest that tomatine is a promising therapeutic candidate for enhancing hepatic glucose metabolism and mitochondrial function in IR-associated metabolic disorders through AMPK activation.
Collapse
Affiliation(s)
| | - Donghwan Kim
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun 55365, Jeonbuk-do, Republic of Korea;
| |
Collapse
|
8
|
Faleti JO, Olasore HSA, Olawale MO, Murtala AA, Banjo TO, Igwo-Ezikpe MN. Association of HindIII Polymorphism of the Lipoprotein Lipase (LPL) Gene (rs320) and Plasma Metabolic Parameters in a Nigerian Population. Biochem Genet 2025:10.1007/s10528-025-11039-w. [PMID: 39899166 DOI: 10.1007/s10528-025-11039-w] [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: 10/04/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025]
Abstract
Genetic variations in the lipoprotein lipase (LPL) gene including the HindIII polymorphism (rs320) have been reported to modify fat metabolism, adiposity, and body weight. However, little attention has been given to the African population. The present study aimed to investigate the relationship between the rs320 gene polymorphism and a number of metabolic and anthropometric parameters in a sample of the Nigerian population. We recruited 236 participants for the study. The participants were required to sign informed consent forms after which information related to their calorie intake and utilization as well as anthropometric measurements were recorded. Plasma metabolic parameters were subsequently determined using an autoanalyzer. Genotyping for HindIII polymorphism was performed using the PCR-RFLP method. The frequencies (n) of T and G alleles were 0.841 (397) and 0.158 (75), while the frequencies (n) of TT, TG, and GG were 0.691(163), 0.301(71), and 0.01(2), respectively. The population was not in Hardy-Weinberg equilibrium (χ2 = 3.717, df = 1, p = 0.841). The anthropometric parameters, the fasting blood glucose, and low-density lipoprotein cholesterol showed no association with the alleles, while plasma high-density lipoprotein cholesterol and total cholesterol were significantly higher among the G allele carriers. However, triglyceride and total protein were significantly higher among the non-G allele carriers. The LPL HindIII gene polymorphism is associated with changes in plasma lipid profile in a sample of the Nigerian population.
Collapse
Affiliation(s)
- Joseph O Faleti
- Department of Biochemistry, College of Medicine, University of Lagos, Idi-Araba Campus, Surulere, Lagos State, Nigeria
| | - Holiness S A Olasore
- Department of Biochemistry, College of Medicine, University of Lagos, Idi-Araba Campus, Surulere, Lagos State, Nigeria.
| | - Matthew O Olawale
- Department of Biochemistry, College of Medicine, University of Lagos, Idi-Araba Campus, Surulere, Lagos State, Nigeria
| | - Abdullahi A Murtala
- Department of Pharmacology and Therapeutics, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria
| | - Taiwo O Banjo
- Department of Medical Microbiology and Parasitology, Obafemi Awolowo College of Health Sciences, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Ogun State, Nigeria
| | - Miriam N Igwo-Ezikpe
- Department of Biochemistry, College of Medicine, University of Lagos, Idi-Araba Campus, Surulere, Lagos State, Nigeria
| |
Collapse
|
9
|
Wang Y, Fu C, Jin H, Yang Y, Li X, Liu K. Lipid metabolism indicators provide tools for the diagnosis of non-alcoholic fatty liver disease: results of a nationwide survey. Front Endocrinol (Lausanne) 2025; 15:1468228. [PMID: 39897962 PMCID: PMC11781989 DOI: 10.3389/fendo.2024.1468228] [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: 07/21/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025] Open
Abstract
Background Cardiometabolic index (CMI), visceral adiposity index (VAI), and lipid accumulation product (LAP) are lipid-related parameters that reflect central obesity, which is closely associated with the development of non-alcoholic fatty liver disease (NAFLD). The aim of this study is to investigate the effectiveness of these lipid-related parameters in diagnosing NAFLD and to compare their predictive abilities. Methods This population-based study extracted datasets from the National Health and Nutrition Examination Survey (NHANES) 2017-2020. CMI, VAI, and LAP were included in the multivariate logistic model as both continuous and categorical variables to assess the relationship between different lipid-related parameters and NAFLD. To further elucidate this connection, we utilized restricted cubic splines and conducted subgroup analysis. Additionally, the receiver operating characteristics (ROC) curve was employed to evaluate the predictive effectiveness of CMI, VAI, and LAP for NAFLD. Results The study included 2,878 adults as the study population, of whom 1,263 participants were diagnosed with NAFLD. When lipid-related parameters were analyzed as continuous variables, they showed a positive correlation with NAFLD. The OR(95%CI) were 2.29(1.81,2.89) for CMI (per 1-unit), 1.40(1.28,1.52) for VAI (per 1-unit) and 1.15(1.11,1.20) for LAP (per 10-units). This correlation remains statistically significant when the lipid-related parameters are analyzed as categorical variables. In descending order of diagnostic capability for NAFLD, the AUC values are as follows: LAP (0.794), CMI (0.752), and VAI (0.719). Conclusion CMI, VAI, and LAP may be important clinical indicators for identifying NAFLD, with LAP demonstrating the best predictive ability among them.
Collapse
Affiliation(s)
- Yongxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chang Fu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hengwei Jin
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yibo Yang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaocong Li
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
- Clinical Trial Research Center, China-Japan Friendship Hospital, Beijing, China
| | - Kai Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Msane S, Khathi A, Sosibo AM. The Effect of the 14:10-Hour Time-Restricted Feeding (TRF) Regimen on Selected Markers of Glucose Homeostasis in Diet-Induced Prediabetic Male Sprague Dawley Rats. Nutrients 2025; 17:292. [PMID: 39861423 PMCID: PMC11768421 DOI: 10.3390/nu17020292] [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: 11/23/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Prediabetes is a condition that often precedes the onset of type 2 diabetes mellitus (T2DM). Literature evidence indicates that prediabetes is reversible, making it an important therapeutic target for preventing the progression to T2DM. Several studies have investigated intermittent fasting as a possible method to manage or treat prediabetes. OBJECTIVES This study evaluated the impact of a 14:10-hour time-restricted feeding (TRF) regimen on leptin concentration, insulin sensitivity and selected markers associated with the insulin signalling pathway and glucose homeostasis in diet-induced prediabetic rats. METHODS Twenty-four male Sprague Dawley rats were obtained and randomly divided into two dietary groups: group 1 (n = 6) received a standard diet and water, while group 2 (n = 18) was provided a high-fat, high-carbohydrate (HFHC) diet supplemented with 15% fructose for a period of 20 weeks to induce prediabetes. After confirming prediabetes, an intermittent fasting (IF) regimen was assigned to the rats while also having untreated and metformin-treated prediabetic rats serving as controls. RESULTS Both IF and HFHC-Met groups yield significantly lower blood glucose, leptin and BMI results compared to the prediabetic group. The IF group yielded significantly lower insulin, HOMA-IR and HbA1C than both controls. CONCLUSIONS The study showed the potential of IF in alleviating prediabetes-induced dysregulation of glucose homeostasis and therefore warrants further investigations into its use in the management of prediabetes.
Collapse
Affiliation(s)
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (S.M.); (A.M.S.)
| | | |
Collapse
|
11
|
Kim JE, Lee JW, Cha GD, Yoon JK. The Potential of Mesenchymal Stem Cell-Derived Exosomes to Treat Diabetes Mellitus. Biomimetics (Basel) 2025; 10:49. [PMID: 39851765 PMCID: PMC11760843 DOI: 10.3390/biomimetics10010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Diabetes mellitus (DM) is a fatal metabolic disease characterized by persistent hyperglycemia. In recent studies, mesenchymal stem cell (MSC)-derived exosomes, which are being investigated clinically as a cell-free therapy for various diseases, have gained attention due to their biomimetic properties that closely resemble natural cellular communication systems. These MSC-derived exosomes inherit the regenerative and protective effects from MSCs, inducing pancreatic β-cell proliferation and inhibiting apoptosis, as well as ameliorating insulin resistance by suppressing the release of various inflammatory cytokines. Consequently, MSC-derived exosomes have attracted attention as a novel treatment for DM as an alternative to stem cell therapy. In this review, we will introduce the potential of MSC-derived exosomes for the treatment of DM by discussing the studies that have used MSC-derived exosomes to treat DM, which have shown therapeutic effects in both type 1 and type 2 DM.
Collapse
Affiliation(s)
| | | | | | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea (G.D.C.)
| |
Collapse
|
12
|
Parraga G, Eddy RL. Power Outage: Mitochondrial Dysfunction in Long COVID Measured Using 1H and 31P MR Spectroscopy. Radiology 2024; 313:e242833. [PMID: 39718501 DOI: 10.1148/radiol.242833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Affiliation(s)
- Grace Parraga
- From the Robarts Research Institute, Department of Medical Biophysics, Division of Respirology, Department of Medicine, and School of Biomedical Engineering, Western University, 1151 Richmond St N, Rm 5235, London, ON, Canada N6A 5B7 (G.P.); and Centre for Heart Lung Innovation, St. Paul's Hospital, BC Children's Hospital Research Institute, Department of Radiology, Department of Pediatrics, University of British Columbia, Vancouver, Canada (R.L.E.)
| | - Rachel L Eddy
- From the Robarts Research Institute, Department of Medical Biophysics, Division of Respirology, Department of Medicine, and School of Biomedical Engineering, Western University, 1151 Richmond St N, Rm 5235, London, ON, Canada N6A 5B7 (G.P.); and Centre for Heart Lung Innovation, St. Paul's Hospital, BC Children's Hospital Research Institute, Department of Radiology, Department of Pediatrics, University of British Columbia, Vancouver, Canada (R.L.E.)
| |
Collapse
|
13
|
Al Harake SN, Abedin Y, Hatoum F, Nassar NZ, Ali A, Nassar A, Kanaan A, Bazzi S, Azar S, Harb F, Ghadieh HE. Involvement of a battery of investigated genes in lipid droplet pathophysiology and associated comorbidities. Adipocyte 2024; 13:2403380. [PMID: 39329369 PMCID: PMC11445895 DOI: 10.1080/21623945.2024.2403380] [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: 12/07/2023] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LDs) are highly specialized energy storage organelles involved in the maintenance of lipid homoeostasis by regulating lipid flux within white adipose tissue (WAT). The physiological function of adipocytes and LDs can be compromised by mutations in several genes, leading to NEFA-induced lipotoxicity, which ultimately manifests as metabolic complications, predominantly in the form of dyslipidemia, ectopic fat accumulation, and insulin resistance. In this review, we delineate the effects of mutations and deficiencies in genes - CIDEC, PPARG, BSCL2, AGPAT2, PLIN1, LIPE, LMNA, CAV1, CEACAM1, and INSR - involved in lipid droplet metabolism and their associated pathophysiological impairments, highlighting their roles in the development of lipodystrophies and metabolic dysfunction.
Collapse
Affiliation(s)
- Sami N. Al Harake
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Yasamin Abedin
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Fatema Hatoum
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Nour Zahraa Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Ali Ali
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Aline Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Sami Azar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| |
Collapse
|
14
|
Shi H, Sim YJ. Effects of weekend-focused exercise on obesity-related hormones and metabolic syndrome markers in male high school students. J Exerc Rehabil 2024; 20:227-234. [PMID: 39781501 PMCID: PMC11704708 DOI: 10.12965/jer.2448632.316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 01/12/2025] Open
Abstract
To examine the changes in obesity-related hormones and metabolic syndrome markers in male high school students with obesity following a weekend-focused moderate- or high-intensity exercise program at the recommended weekly physical activity level, or a program of regular exercise 3 times a week at moderate intensity, over a 10-week period. Forty-eight male high school students who were obese with a body fat percentage of ≥25% were randomly assigned to one of three groups: a regular moderate-intensity exercise group (n=17) that freely selected and performed moderate-intensity aerobic and resistance training exercises, every Monday, Wednesday, and Friday, for a total of 150-300 min/wk; a weekend-focused moderate-intensity exercise group (n=15) that freely selected and performed aerobic and resistance training exercises every Saturday for 150-300 min; and a week-end-focused high-intensity exercise group (n=16) that freely selected and performed aerobic and resistance training exercises every Sunday for 75-150 min. Insulin and leptin levels significantly decreased in all the groups, with the greatest reduction in the regular exercise group. Abdominal circumference and triglyceride levels significantly decreased in all the groups. Fasting glucose decreased only in the regular exercise group. High-density lipoprotein cholesterol significantly increased in both the regular and weekend-focused moderate-intensity exercise groups. No significant differences in adiponectin levels, and systolic and diastolic blood pressure were observed between the groups. A weekend-focused exercise program has health effects similar to those of regular exercise, highlighting the importance of meeting the recommended weekly physical activity levels.
Collapse
Affiliation(s)
- Haoyu Shi
- Department of Physical Education, Kunsan National University, Gunsan,
Korea
| | - Young-Je Sim
- Department of Physical Education, Kunsan National University, Gunsan,
Korea
| |
Collapse
|
15
|
Ullman JC, Dick RA, Linzner D, Minga T, Tep S, Satterfield TF, Xi Y, Beattie DT, Marmon T, Neutel JM, Chung B, Leeds JM, Noonberg SB, Green EM, Bernstein HS. First-in-Human Evaluation of Safety, Pharmacokinetics and Muscle Glycogen Lowering of a Novel Glycogen Synthase 1 Inhibitor for the Treatment of Pompe Disease. Clin Pharmacol Ther 2024; 116:1580-1592. [PMID: 39439155 DOI: 10.1002/cpt.3470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Pompe disease is a rare glycogen storage disease caused by mutations in the enzyme acid α-glucosidase (GAA) resulting in pathological accumulation of glycogen in muscle tissues leading to progressive weakness and respiratory dysfunction. Enzyme replacement therapy (ERT) with GAA is currently the sole treatment option for patients with Pompe disease. ERT burdens patients with frequent intravenous infusions while insufficiently halting disease progression due to incomplete ERT skeletal muscle distribution. Glycogen synthase 1 (GYS1) has been proposed as a substrate reduction therapy (SRT) target for Pompe disease. Here, we report results from the first-in-human study of the orally available GYS1 inhibitor MZE001 in healthy subjects. In 88 participants, MZE001 was well-tolerated up to a single dose of 480 mg BID and multiple doses of 720 mg BID for 10 days. Noncompartmental analysis determined that the half-life and Ctrough concentrations of MZE001 could provide efficacious exposures with once or twice daily oral dosing. Change from baseline of peripheral blood mononuclear cell (PBMC) glycogen, which correlated with muscle glycogen levels in preclinical models, was significantly reduced dose-dependently following 10 days of MZE001 treatment in healthy subjects. A muscle biopsy sub-study demonstrated that 10 days of MZE001 (480 mg BID) dosing safely and substantially lowered muscle glycogen stores in healthy adults. This correlated with the PBMC exposure response and supports the use of PBMC glycogen reduction as a surrogate for muscle response, and MZE001 potential for development as the first oral substrate reduction therapy for patients with Pompe disease.
Collapse
Affiliation(s)
| | - Ryan A Dick
- Maze Therapeutics, South San Francisco, California, USA
| | | | - Todd Minga
- Maze Therapeutics, South San Francisco, California, USA
| | - Samnang Tep
- Maze Therapeutics, South San Francisco, California, USA
| | | | - Yannan Xi
- Maze Therapeutics, South San Francisco, California, USA
| | | | | | - Joel M Neutel
- Orange County Research Center, Tustin, California, USA
| | - Bernard Chung
- Maze Therapeutics, South San Francisco, California, USA
| | - Janet M Leeds
- Maze Therapeutics, South San Francisco, California, USA
| | | | - Eric M Green
- Maze Therapeutics, South San Francisco, California, USA
| | | |
Collapse
|
16
|
Lee S, Kim HK. α-Tocopherol and γ-tocopherol decrease inflammatory response and insulin resistance during the interaction of adipocytes and macrophages. Nutr Res Pract 2024; 18:761-773. [PMID: 39651320 PMCID: PMC11621430 DOI: 10.4162/nrp.2024.18.6.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/10/2024] [Accepted: 08/08/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND/OBJECTIVES The infiltration of macrophages into adipose tissue mediates chronic inflammation that is associated with insulin resistance in obesity. Although vitamin E is beneficial against insulin resistance, its impact on adipose tissue inflammation has not been elucidated. This study aims to investigate the effects of α-tocopherol and γ-tocopherol, major vitamin E isoforms, on the interaction between macrophages and adipocytes with regard to obesity-induced inflammation and insulin resistance. MATERIALS/METHODS Hypertrophied 3T3-L1 adipocytes were cocultured with RAW 264.7 macrophages and treated with α-tocopherol or γ-tocopherol at 12.5, 25, and 50 µM. The inflammatory cytokines (monocyte chemoattractant protein-1, tumor necrosis factor-α, and interleukin-6) and free fatty acid (FFA) release were measured by assay kits, and nuclear factor-kappaB (NF-κB) and c-Jun NH2 terminal kinase (JNK) signals were evaluated by immunoblotting. Glucose uptake was measured with a fluorescent glucose derivative. RESULTS Treatment with α-tocopherol and γ-tocopherol restrained the coculture-induced increase in cytokines and FFA release. γ-Tocopherol exhibited greater suppression of inflammatory cytokines at 12.5 and 25 µM (P < 0.001). Both tocopherols inhibited NF-κB activation by limiting translocation of NF-κB (p65) to the nucleus, with γ-tocopherol showing a stronger effect compared to α-tocopherol. α-Tocopherol inhibited JNK phosphorylation at 50 μM, whereas γ-tocopherol did not. Furthermore, coculture with macrophages impaired glucose uptake in response to insulin, but both tocopherols restored insulin responsiveness (P < 0.01). CONCLUSION α-Tocopherol and γ-tocopherol effectively mitigate inflammation induced by adipocyte-macrophage interaction, thereby ameliorating coculture-induced insulin resistance. These findings suggest the therapeutic potential of tocopherols in managing obesity-related metabolic dysfunction.
Collapse
Affiliation(s)
- Sella Lee
- Department of Food Science and Nutrition, The Catholic University of Korea, Bucheon 14662, Korea
| | - Hye-Kyeong Kim
- Department of Food Science and Nutrition, The Catholic University of Korea, Bucheon 14662, Korea
| |
Collapse
|
17
|
Lv X, Nie C, Shi Y, Qiao Q, Gao J, Zou Y, Yang J, Chen L, Hou X. Ergothioneine ameliorates metabolic dysfunction-Associated Steatotic Liver Disease (MASLD) by enhancing autophagy, inhibiting oxidative damage and inflammation. Lipids Health Dis 2024; 23:395. [PMID: 39609792 PMCID: PMC11604011 DOI: 10.1186/s12944-024-02382-9] [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: 04/16/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatosis liver disease (MASLD) is one of the most common metabolic liver diseases around the world, whose prevalence continues to increase. Currently, there are few medications to treat MASLD. Ergothioneine is a natural compound derived from mushrooms whose sulfhydryl groups confer unique antioxidant, anti-inflammatory and detoxifying effects. Currently, research on the therapeutic effects of ergothioneine in MASLD is unknown. Therefore, this study explored the effect and mechanism of EGT in MASLD. METHODS The ameliorative effects and mechanisms of ergothioneine on MASLD were evaluated using HFD mice and PA-treated AML12 cells. Mouse body weight, body fat, IPGTT, IPITT, immunohistochemistry, serum biochemical indices, and staining of liver sections were assayed to verify the protective role of ergothioneine in MASLD. RNA-seq was applied to explore the mechanism of action of ergothioneine. The role of ergothioneine in AML12 was confirmed by western blotting, qPCR, ELISA, Oil Red O staining, flow cytometry, and ROS assays. Subsequently, the 3-methyladenine (3-MA, an autophagy inhibitor) was subsequently used to confirm that ergothioneine alleviated MASLD by promoting autophagy. RESULTS Ergothioneine reduced body weight, body fat and blood lipids, and improved insulin resistance and lipid and glycogen deposition in MASLD mice. Furthermore, ergothioneine was found to increase autophagy levels and attenuate oxidative damage, inflammation, and apoptosis. In contrast, intervention with 3-MA abrogated these effects, suggesting that ergothioneine ameliorated effects by promoting autophagy. CONCLUSION Ergothioneine may be a drug with great therapeutic potential for MASLD. Furthermore, this protective effect was mediated through the activation of autophagy.
Collapse
Affiliation(s)
- Xiaoyu Lv
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Chenyu Nie
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Yihan Shi
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Qincheng Qiao
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Jing Gao
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Ying Zou
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Jingwen Yang
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
| | - Li Chen
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, Shandong, 250012, China
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong, 250012, China
| | - Xinguo Hou
- Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, 107 Wenhuaxi Road, Li Xia district, Jinan, Shandong, 250012, China.
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, Shandong, 250012, China.
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong, 250012, China.
| |
Collapse
|
18
|
Alic L, Dendinovic K, Papac-Milicevic N. The complement system in lipid-mediated pathologies. Front Immunol 2024; 15:1511886. [PMID: 39635529 PMCID: PMC11614835 DOI: 10.3389/fimmu.2024.1511886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
The complement system, a coordinator and facilitator of the innate immune response, plays an essential role in maintaining host homeostasis. It promotes clearance of pathogen- and danger-associated molecular patterns, regulates adaptive immunity, and can modify various metabolic processes such as energy expenditure, lipid metabolism, and glucose homeostasis. In this review, we will focus on the intricate interplay between complement components and lipid metabolism. More precisely, we will display how alterations in the activation and regulation of the complement system affect pathological outcome in lipid-associated diseases, such as atherosclerosis, obesity, metabolic syndrome, age-related macular degeneration, and metabolic dysfunction-associated steatotic liver disease. In addition to that, we will present and evaluate underlying complement-mediated physiological mechanisms, observed both in vitro and in vivo. Our manuscript will demonstrate the clinical significance of the complement system as a bridging figure between innate immunity and lipid homeostasis.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Kristina Dendinovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Suder A, Makiel K, Targosz A, Kosowski P, Malina RM. Effects of exercise and dietary interventions on asprosin, leptin, and lipid metabolism in males with abdominal obesity, a randomized controlled trial. Sci Rep 2024; 14:28109. [PMID: 39548289 PMCID: PMC11568226 DOI: 10.1038/s41598-024-79853-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
Addressing abdominal obesity requires multifaceted strategies, with physical activity and diet playing a pivotal role. The objective of this study was to assess alterations in body composition, adipokine concentrations, insulin resistance parameters, and lipid metabolism in males with abdominal obesity following two distinct interventions: exercise alone and exercise combined with a specific diet. The study involved 44 males with abdominal obesity (average age 34.7 ± 5.5 years, waist circumference [WC] 110.3 ± 8.5), randomly assigned to three groups: an experimental group with aerobic-resistance exercise (EG, n = 16), an experimental group with aerobic-resistance exercise combined with a high-protein, low-glycemic index carbohydrate diet (EDG, n = 16), both interventions lasting 6 weeks, and a control group without interventions (CG, n = 12). Body composition (body mass [BM], body fat percentage [BF%], fat-free mass [FFM], android body fat percentage [ANDR]), as well as biochemical blood analyses (asprosin [ASP], leptin [LEP], quantitative insulin sensitivity check index [QUICKI], and total cholesterol [TC]), were conducted at baseline and after 6 weeks of intervention. The impact of interventions on the analyzed variables among groups was assessed using mixed ANOVA tests with post-hoc comparisons. Effect size (ES) was also evaluated using 𝜂p2. Significant reductions in ASP concentration after intervention were observed in both EG (p = 0.04) and EDG (p = 0.01). However, post-hoc tests revealed a decrease in LEP only in the EDG group (p < 0.01). In EDG substantial decreases after 6 weeks of intervention were noted in BM (p < 0.01), BF% (p < 0.01), ANDR (p < 0.01) and TC (p < 0.01). The most notable increase in FFM was observed in the EDG group (p < 0.01). More favourable metabolic outcomes were confirmed in the group combining diet with exercise, where there was a notable reduction in ASP levels by 16% and LEP by 48% after 6 weeks of intervention, compared to the group undergoing exercise alone.
Collapse
Affiliation(s)
- Agnieszka Suder
- Department of Anatomy, Faculty of Physical Rehabilitation, University of Physical Education, Cracow, 31-571, Poland.
| | - Karol Makiel
- Department of Anatomy, Faculty of Physical Rehabilitation, University of Physical Education, Cracow, 31-571, Poland
| | - Aneta Targosz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, 31-531, Poland
| | - Piotr Kosowski
- Department of Petroleum Engineering, AGH University, Cracow, 30-059, Poland
| | - Robert M Malina
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, TX, 78712, USA
- School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| |
Collapse
|
20
|
Wang Y, Wang X, Zeng L. Lipid Accumulation Product as a Predictor of Prediabetes and Diabetes: Insights From NHANES Data (1999-2018). J Diabetes Res 2024; 2024:2874122. [PMID: 39559713 PMCID: PMC11573446 DOI: 10.1155/2024/2874122] [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: 12/02/2023] [Revised: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
Background: The study investigates the association between lipid accumulation product (LAP) and the risk of prediabetes and diabetes. LAP, a measure indicating lipid overaccumulation, is hypothesized to be a significant predictor for these conditions. This research utilizes data from the National Health and Nutrition Examination Survey (NHANES) conducted between 1999 and 2018. Methods: The study followed a structured methodology, starting with data extraction from the NHANES database. Participants' eligibility was determined based on specific inclusion and exclusion criteria, resulting in a final sample size of 24,121 individuals. LAP was calculated using established formulas for men and women. The diagnosis of prediabetes and diabetes was based on standard medical criteria, including HbA1c levels, fasting plasma glucose, and oral glucose tolerance test (OGTT) results. Covariates like demographic variables, lifestyle factors, and other health indicators were also considered. Statistical analysis involved categorizing LAP into quartiles and employing logistic regression models to examine the relationship between LAP and the risk of prediabetes and diabetes. Results: Participants in the highest LAP quartile exhibited distinct characteristics: older age, lower education levels, more former smokers and drinkers, higher blood pressure and cholesterol levels, and greater use of medications. A positive association was observed between LAP and the incidence of prediabetes and diabetes across all models. Specifically, each 10-unit increase in LAP was linked to a 22% increase in risk. Nonlinear relationships were also explored, revealing an inflection point in the risk correlation at an LAP value of 68.1. Conclusion: The study concludes that LAP is a significant predictor of prediabetes and diabetes risk, with higher LAP levels correlating with increased risk. This finding underscores the potential of LAP as a useful marker in identifying individuals at higher risk for these conditions. It also highlights the importance of considering LAP in preventive health strategies.
Collapse
Affiliation(s)
- Yan Wang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| | - Xiaolan Wang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| | - Ling Zeng
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Bergman BC, Zemski Berry K, Garfield A, Keller A, Zarini S, Bowen S, McKenna C, Kahn D, Pavelka J, Macias E, Uhlson C, Johnson C, Russ HA, Viesi CH, Seldin M, Liu C, Doliba N, Schoen J, Rothchild K, Hazel K, Naji A. Human peripancreatic adipose tissue paracrine signaling impacts insulin secretion, blood flow, and gene transcription. J Clin Endocrinol Metab 2024:dgae767. [PMID: 39484843 DOI: 10.1210/clinem/dgae767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/14/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024]
Abstract
CONTEXT Adipose tissue accumulation around non-adipose tissues is associated with obesity and metabolic disease. One relatively unstudied depot is peripancreatic adipose tissue (PAT) that accumulates in obesity and insulin resistance and may impact beta cell function. Pancreatic lipid accumulation and PAT content are negatively related to metabolic outcomes in humans, but these studies are limited by the inability to pursue mechanisms. OBJECTIVE We obtained PAT from human donors through the Human Pancreas Analysis Program to evaluate differences in paracrine signaling compared to subcutaneous adipose tissue (SAT), as well as effects of the PAT secretome on aortic vasodilation, human islet insulin secretion, and gene transcription using RNAseq. RESULTS PAT had greater secretion of IFN-γ and most inflammatory eicosanoids compared to SAT. Secretion of adipokines negatively related to metabolic health were also increased in PAT compared to SAT. We found no overall effects of PAT compared to SAT on human islet insulin secretion, however, insulin secretion was suppressed after PAT exposure from men compared to women. Vasodilation was significantly dampened by PAT conditioned media, an effect explained almost completely by PAT from men and not women. Islets treated with PAT showed selective changes in lipid metabolism pathways while SAT altered cellular signaling and growth. RNAseq analysis showed changes in islet gene transcription impacted by PAT compared to SAT, with the biggest changes found between PAT based on sex. CONCLUSION The PAT secretome is metabolically negative compared to SAT, and impacts islet insulin secretion, blood flow, and gene transcription in a sex dependent manner.
Collapse
Affiliation(s)
- Bryan C Bergman
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karin Zemski Berry
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amanda Garfield
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amy Keller
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | - Simona Zarini
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sophia Bowen
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Colleen McKenna
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Darcy Kahn
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jay Pavelka
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Emily Macias
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Charis Uhlson
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chris Johnson
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Holger A Russ
- College of Medicine, Department of Pharmacology and Therapeutics, University of Florida USA
- Diabetes Institute, University of Florida USA
| | - Carlos H Viesi
- Department of Biological Chemistry and the Center for Epigenetics and Metabolism, University of California, Irvine, CA, USA
| | - Marcus Seldin
- Department of Biological Chemistry and the Center for Epigenetics and Metabolism, University of California, Irvine, CA, USA
| | - Chengyang Liu
- University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Nicolai Doliba
- University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Jonathan Schoen
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kevin Rothchild
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kweku Hazel
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ali Naji
- University of Pennsylvania Medical Center, Philadelphia, PA, USA
| |
Collapse
|
22
|
Xu W, Zhang D, Ma Y, Gaspar RC, Kahn M, Nasiri A, Murray S, Samuel VT, Shulman GI. Ceramide synthesis inhibitors prevent lipid-induced insulin resistance through the DAG-PKCε-insulin receptor T1150 phosphorylation pathway. Cell Rep 2024; 43:114746. [PMID: 39302831 DOI: 10.1016/j.celrep.2024.114746] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/22/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
Inhibition of the ceramide synthetic pathway with myriocin or an antisense oligonucleotide (ASO) targeting dihydroceramide desaturase (DES1) both improved hepatic insulin sensitivity in rats fed either a saturated or unsaturated fat diet and was associated with reductions in both hepatic ceramide and plasma membrane (PM)-sn-1,2-diacylglycerol (DAG) content. The insulin sensitizing effects of myriocin and Des1 ASO were abrogated by acute treatment with an ASO against DGAT2, which increased hepatic PM-sn-1,2-DAG but not hepatic C16 ceramide content. Increased PM-sn-1,2-DAG content was associated with protein kinase C (PKC)ε activation, increased insulin receptor (INSR)T1150 phosphorylation leading to reduced insulin-stimulated INSRY1152/AktS473 phosphorylation, and impaired insulin-mediated suppression of endogenous glucose production. These results demonstrate that inhibition of de novo ceramide synthesis by either myriocin treatment or DES1 knockdown protects against lipid-induced hepatic insulin resistance through a C16 ceramide-independent mechanism and that they mediate their effects to protect from lipid-induced hepatic insulin resistance via the PM-sn-1,2-DAG-PKCε-INSRT1150 phosphorylation pathway.
Collapse
Affiliation(s)
- Weiwei Xu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Endocrinology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yumin Ma
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Endocrinology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Rafael C Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mario Kahn
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ali Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sue Murray
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA
| | - Varman T Samuel
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
23
|
Saxena A, Tiwari P, Gupta S, Mandia R, Banshiwal RC, Lamoria RK, Anjana RM, Radha V, Mohan V, Mathur SK. Exploring lipodystrophy gene expression in adipocytes: unveiling insights into the pathogenesis of insulin resistance, type 2 diabetes, and clustering diseases (metabolic syndrome) in Asian Indians. Front Endocrinol (Lausanne) 2024; 15:1468824. [PMID: 39444451 PMCID: PMC11496143 DOI: 10.3389/fendo.2024.1468824] [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: 07/22/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Background Studying the molecular mechanisms of lipodystrophy can provide valuable insights into the pathophysiology of insulin resistance (IR), type 2 diabetes (T2D), and other clustering diseases [metabolic syndrome (MetS)] and its underlying adipocentric disease (MetS disease). Methods A high-confidence lipodystrophy gene panel comprising 50 genes was created, and their expressions were measured in the visceral and subcutaneous (both peripheral and abdominal) adipose depots of MetS and non-MetS individuals at a tertiary care medical facility. Results Most lipodystrophy genes showed significant downregulation in MetS individuals compared to non-MetS individuals in both subcutaneous and visceral depots. In the abdominal compartment, all the genes showed relatively higher expression in visceral depot as compared to their subcutaneous counterpart, and this difference narrowed with increasing severity of MetS. Their expression level shows an inverse correlation with T2D, MetS, and HOMA-IR and with other T2D-related intermediate traits. Results also demonstrated that individualization of MetS patients could be done based on adipose tissue expression of just 12 genes. Conclusion Adipose tissue expression of lipodystrophy genes shows an association with MetS and its intermediate phenotypic traits. Mutations of these genes are known to cause congenital lipodystrophy syndromes, whereas their altered expression in adipose tissue contributes to the pathogenesis of IR, T2D, and MetS.
Collapse
Affiliation(s)
- Aditya Saxena
- Department of Computer Engineering & Applications, GLA University, Mathura, India
| | - Pradeep Tiwari
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Jaipur, India
| | - Shalu Gupta
- Department of General Surgery, Sawai Man Singh (SMS) Medical College and Attached Hospital, Jaipur, India
| | - Rajendra Mandia
- Department of General Surgery, Sawai Man Singh (SMS) Medical College and Attached Hospital, Jaipur, India
| | - Ramesh C. Banshiwal
- Department of Orthopedics, Sawai Man Singh (SMS) Medical College and Attached Hospital, Jaipur, India
| | - Ravinder Kumar Lamoria
- Department of Orthopedics, Sawai Man Singh (SMS) Medical College and Attached Hospital, Jaipur, India
| | - Ranjit Mohan Anjana
- Department of Diabetology, Madras Diabetes Research Foundation, Chennai, Tamil Nadu, India
| | - Venkatesan Radha
- Department of Diabetology, Madras Diabetes Research Foundation, Chennai, Tamil Nadu, India
| | - Viswanathan Mohan
- Department of Diabetology, Madras Diabetes Research Foundation, Chennai, Tamil Nadu, India
| | - Sandeep Kumar Mathur
- Department of Endocrinology, Sawai Man Singh (SMS) Medical College, Jaipur, India
| |
Collapse
|
24
|
Fermaintt CS, Wacker SA. Malate dehydrogenase as a multi-purpose target for drug discovery. Essays Biochem 2024; 68:147-160. [PMID: 38818725 DOI: 10.1042/ebc20230081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024]
Abstract
Malate dehydrogenase (MDH) enzymes play critical roles in cellular metabolism, facilitating the reversible conversion of malate to oxaloacetate using NAD+/NADH as a cofactor. The two human isoforms of MDH have roles in the citric acid cycle and the malate-aspartate shuttle, and thus both are key enzymes in aerobic respiration as well as regenerating the pool of NAD+ used in glycolysis. This review highlights the potential of MDH as a therapeutic drug target in various diseases, including metabolic and neurological disorders, cancer, and infectious diseases. The most promising molecules for targeting MDH have been examined in the context of human malignancies, where MDH is frequently overexpressed. Recent studies have led to the identification of several antagonists, some of which are broad MDH inhibitors while others have selectivity for either of the two human MDH isoforms. Other promising compounds have been studied in the context of parasitic MDH, as inhibiting the function of the enzyme could selectively kill the parasite. Research is ongoing with these chemical scaffolds to develop more effective small-molecule drug leads that would have great potential for clinical applications.
Collapse
Affiliation(s)
- Charles S Fermaintt
- Department of Chemistry and Biochemistry, University of the Incarnate Word, San Antonio, TX, U.S.A
| | - Sarah A Wacker
- Department of Chemistry and Biochemistry, Manhattan College, The Bronx, NY, U.S.A
| |
Collapse
|
25
|
Gaspar RC, Sakuma I, Nasiri A, Hubbard BT, LaMoia TE, Leitner BP, Tep S, Xi Y, Green EM, Ullman JC, Petersen KF, Shulman GI. Small molecule inhibition of glycogen synthase I reduces muscle glycogen content and improves biomarkers in a mouse model of Pompe disease. Am J Physiol Endocrinol Metab 2024; 327:E524-E532. [PMID: 39171753 PMCID: PMC11482269 DOI: 10.1152/ajpendo.00175.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
Abstract
Pompe disease is a rare genetic disorder caused by a deficiency of the enzyme acid alpha-glucosidase (GAA). This enzyme is responsible for breaking down glycogen, leading to the abnormal accumulation of glycogen, which results in progressive muscle weakness and metabolic dysregulation. In this study, we investigated the hypothesis that the small molecule inhibition of glycogen synthase I (GYS1) may reduce muscle glycogen content and improve metabolic dysregulation in a mouse model of Pompe disease. To address this hypothesis, we studied four groups of male mice: a control group of wild-type (WT) B6129SF1/J mice fed either regular chow or a GYS1 inhibitor (MZ-101) diet (WT-GYS1), and Pompe model mice B6;129-Gaatm1Rabn/J fed either regular chow (GAA-KO) or MZ-101 diet (GAA-GYS1) for 7 days. Our findings revealed that GAA-KO mice exhibited abnormal glycogen accumulation in the gastrocnemius, heart, and diaphragm. In contrast, inhibiting GYS1 reduced glycogen levels in all tissues compared with GAA-KO mice. Furthermore, GAA-KO mice displayed reduced spontaneous activity during the dark cycle compared with WT mice, whereas GYS1 inhibition counteracted this effect. Compared with GAA-KO mice, GAA-GYS1 mice exhibited improved glucose tolerance and whole body insulin sensitivity. These improvements in insulin sensitivity could be attributed to increased AMP-activated protein kinase phosphorylation in the gastrocnemius of WT-GYS1 and GAA-GYS1 mice. Additionally, the GYS1 inhibitor led to a reduction in the phosphorylation of GSS641 and the LC3 autophagy marker. Together, our results suggest that targeting GYS1 could serve as a potential strategy for treating glycogen storage disorders and metabolic dysregulation.NEW & NOTEWORTHY We investigated the effects of small molecule inhibition of glycogen synthase I (GYS1) on glucose metabolism in a mouse model of Pompe disease. GYS1 inhibition reduces abnormal glycogen accumulation and molecular biomarkers associated with Pompe disease while also improving glucose intolerance. Our results collectively demonstrate that the GYS1 inhibitor represents a novel approach to substrate reduction therapy for Pompe disease.
Collapse
Affiliation(s)
- Rafael Calais Gaspar
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Ikki Sakuma
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Ali Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Brandon T Hubbard
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Traci E LaMoia
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Brooks P Leitner
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Samnang Tep
- Maze Therapeutics, South San Francisco, California,United States
| | - Yannan Xi
- Maze Therapeutics, South San Francisco, California,United States
| | - Eric M Green
- Maze Therapeutics, South San Francisco, California,United States
| | - Julie C Ullman
- Maze Therapeutics, South San Francisco, California,United States
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, United States
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, United States
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States
| |
Collapse
|
26
|
Yadav M, Verma S, Tiwari P, Mugale MN. Unraveling the mechanisms of hepatogenous diabetes and its therapeutic perspectives. Life Sci 2024; 353:122934. [PMID: 39089644 DOI: 10.1016/j.lfs.2024.122934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
The review focused mainly on the pathogenesis of hepatogenous diabetes (HD) in liver cirrhosis (LC). This review reveals parallels between the mechanisms of metabolic dysfunction observed in LC and type II diabetes (T2DM), suggesting a shared pathway leading to HD. It underscores the role of insulin in HD pathogenesis, highlighting key factors such as insulin signaling, glucose metabolism, insulin resistance (IR), and the influence of adipocytes. Furthermore, the impact of adipose tissue accumulation, fatty acid metabolism, and pro-inflammatory cytokines like Tumor necrosis factor-α (TNF-α) on IR are discussed in the context of HD. Altered signaling pathways, disruptions in the endocrine system, liver inflammation, changes in muscle mass and composition, and modifications to the gut microbiota collectively contribute to the complex interplay linking cirrhosis and HD. This study highlights how important it is to identify and treat this complex condition in cirrhotic patients by thoroughly analyzing the link between cirrhosis, IR, and HD. It also emphasizes the vitality of targeted interventions. Cellular and molecular investigations into IR have revealed potential therapeutic targets for managing and preventing HD.
Collapse
Affiliation(s)
- Manisha Yadav
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Smriti Verma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Purnima Tiwari
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
27
|
Beddows CA, Shi F, Horton AL, Dalal S, Zhang P, Ling CC, Yong VW, Loh K, Cho E, Karagiannis C, Rose AJ, Montgomery MK, Gregorevic P, Watt MJ, Packer NH, Parker BL, Brown RM, Moh ESX, Dodd GT. Pathogenic hypothalamic extracellular matrix promotes metabolic disease. Nature 2024; 633:914-922. [PMID: 39294371 PMCID: PMC11424483 DOI: 10.1038/s41586-024-07922-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Metabolic diseases such as obesity and type 2 diabetes are marked by insulin resistance1,2. Cells within the arcuate nucleus of the hypothalamus (ARC), which are crucial for regulating metabolism, become insulin resistant during the progression of metabolic disease3-8, but these mechanisms are not fully understood. Here we investigated the role of a specialized chondroitin sulfate proteoglycan extracellular matrix, termed a perineuronal net, which surrounds ARC neurons. In metabolic disease, the perineuronal net of the ARC becomes augmented and remodelled, driving insulin resistance and metabolic dysfunction. Disruption of the perineuronal net in obese mice, either enzymatically or with small molecules, improves insulin access to the brain, reversing neuronal insulin resistance and enhancing metabolic health. Our findings identify ARC extracellular matrix remodelling as a fundamental mechanism driving metabolic diseases.
Collapse
Affiliation(s)
- Cait A Beddows
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Feiyue Shi
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anna L Horton
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sagar Dalal
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Ping Zhang
- Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kim Loh
- St Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria, Australia
| | - Chris Karagiannis
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Adam J Rose
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Neurology, The University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicolle H Packer
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robyn M Brown
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Edward S X Moh
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Garron T Dodd
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
28
|
Andrews RR, Anderson KR, Fry JL. Sex-Specific Variation in Metabolic Responses to Diet. Nutrients 2024; 16:2921. [PMID: 39275236 PMCID: PMC11397081 DOI: 10.3390/nu16172921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
Suboptimal nutrition is a leading cause of cardiometabolic disease and mortality. Biological sex is a variable that influences individual responses to dietary components and may modulate the impact of diet on metabolic health and disease risk. This review describes findings of studies reporting how biological sex may associate with or affect metabolic outcomes or disease risk in response to varying dietary macronutrient content, Mediterranean diet, Western diet, and medical very low-calorie diet. Although few dietary interventions have been specifically designed to identify sex-diet interactions, future studies improving understanding how sex influences dietary responses could inform precision nutrition interventions for disease prevention and management.
Collapse
Affiliation(s)
- Reya R Andrews
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Kayla R Anderson
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Jean L Fry
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
29
|
Koshta K, Chauhan A, Singh S, Srivastava V. Prenatal arsenic exposure alters EZH2-H3K27me3 occupancy at TNF-α promoter leading to insulin resistance and metabolic syndrome in a mouse model. ENVIRONMENT INTERNATIONAL 2024; 190:108929. [PMID: 39098089 DOI: 10.1016/j.envint.2024.108929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/29/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
The global prevalence of Metabolic Syndrome (MetS) is continuously rising and exposure to environmental toxicants such as arsenic could be contributing to this rapid surge. In this study, we have assessed the effects of prenatal arsenic exposure on insulin resistance and MetS parameters in a mouse model, and an underlying mechanism was identified. We found that prenatal arsenic exposure promotes insulin resistance and adipocyte dysfunction which leads to the early onset of MetS in male offspring. Primary adipocytes isolated from 20-week-old arsenic-exposed offspring showed hypertrophy, elevated basal lipolysis, and impaired insulin response along with enhanced expression of Tumor necrosis factor-alpha (TNF-α). TNF-α levels were consistently high at gestational day 15.5 (GD15.5) as well as primary adipocytes of 6-week-old arsenic-exposed male offspring. Along with TNF-α, downstream p-JNK1/2 levels were also increased, which led to inhibitory phosphorylation of IRS1and reduced GLUT4 translocation upon insulin stimulation in adipocytes. Insulin response and downstream signaling were restored upon TNF-α inhibition, confirming its central role. The persistent overexpression of TNF-α in adipocytes of arsenic-exposed mice resulted from diminished EZH2 occupancy and reduced H3K27me3 (gene silencing histone marks) at the TNF-α promoter. This further led to chromatin relaxation, recruitment of c-Jun and CBP/p300, formation of an enhanceosome complex, and TNF-α expression. Our findings show how prenatal arsenic exposure can epigenetically modulate TNF-α expression to promote adipocyte dysfunction and insulin resistance which contributes to the early onset of MetS in offspring.
Collapse
Affiliation(s)
- Kavita Koshta
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Vishvigyan Bhawan, Mahatma Gandhi Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anchal Chauhan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Vishvigyan Bhawan, Mahatma Gandhi Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sukhveer Singh
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Vishvigyan Bhawan, Mahatma Gandhi Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vikas Srivastava
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Vishvigyan Bhawan, Mahatma Gandhi Marg, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
30
|
Cao C, Wei C, Han Y, Luo J, Xi P, Chen J, Xiao X, Hu H, Qi D. Association between excessive alcohol consumption and incident diabetes mellitus among Japanese based on propensity score matching. Sci Rep 2024; 14:17274. [PMID: 39068183 PMCID: PMC11283479 DOI: 10.1038/s41598-024-68202-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
The available evidence on the connection between excessive alcohol consumption and diabetes is controversial. Therefore, the primary objective of this investigation was to examine the connection between excessive alcohol consumption and incident diabetes in a Japanese population through the utilization of propensity score matching (PSM) analysis. Our retrospective cohort study encompassed a sample of 15,464 Japanese individuals who were initially free of diabetes between the years 2004 and 2015. The study utilized comprehensive medical records of individuals who underwent a physical examination. Employing a one:one PSM analysis, the current research included 2298 individuals with and without excessive alcohol consumption. Furthermore, a doubly robust estimation method was employed to ascertain the connection between excessive alcohol consumption and diabetes. The findings revealed that individuals with excessive alcohol consumption exhibited a 73% higher likelihood of developing diabetes (HR = 1.73, 95% CI 1.08-2.77). Furthermore, upon adjusting for variables, the PSM cohort demonstrated that individuals with excessive alcohol consumption had a 78% increased risk of developing diabetes in comparison to those with non-excessive alcohol consumption (HR = 1.78, 95% CI 1.08-2.93). Individuals with excessive alcohol consumption were found to have a 73% higher risk of developing diabetes compared to those with non-excessive alcohol consumption, even after controlling for propensity score (HR = 1.73, 95% CI 1.08-2.78). Participants in the PSM cohort with excessive alcohol consumption had a 73% higher risk of developing diabetes than those with non-excessive alcohol consumption after controlling for confounding factors. These findings underscore the importance of alcohol consumption guidelines aimed at reducing excessive drinking. Clinicians should be vigilant in screening for alcohol use in patients, particularly those at risk for diabetes, and provide appropriate counseling and resources to support alcohol reduction.
Collapse
Affiliation(s)
- Changchun Cao
- Department of Rehabilitation, Shenzhen Dapeng New District Nan'ao People's Hospital, Shenzhen, 518000, Guangdong Province, China
| | - Cuimei Wei
- Department of Geriatrics, Futian District, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Shenzhen, 518000, Guangdong Province, China
| | - Yong Han
- Department of Emergency, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518000, Guangdong Province, China
| | - Jiao Luo
- Department of Rehabilitation, Shenzhen Dapeng New District Nan'ao People's Hospital, Shenzhen, 518000, Guangdong Province, China
| | - Ping Xi
- Department of Geriatrics, Futian District, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Shenzhen, 518000, Guangdong Province, China
| | - Jingying Chen
- Department of Geriatrics, Futian District, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Shenzhen, 518000, Guangdong Province, China
| | - Xiaohua Xiao
- Department of Geriatrics, Futian District, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Shenzhen, 518000, Guangdong Province, China.
| | - Haofei Hu
- Department of Nephrology, Futian District, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Shenzhen, 518000, Guangdong Province, China.
| | - Dongli Qi
- Department of Nephrology, Futian District, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No.3002, Sungang West Road, Shenzhen, 518000, Guangdong Province, China.
| |
Collapse
|
31
|
Kandror KV. Self-assembly of the insulin-responsive vesicles creates a signaling platform for the insulin action on glucose uptake. VITAMINS AND HORMONES 2024; 128:93-121. [PMID: 40097254 DOI: 10.1016/bs.vh.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
In fat and skeletal muscle cells, insulin causes plasma membrane translocation of specialized insulin-responsive vesicles, or IRVs. These vesicles consist of multiple copies of Glut4, sortilin, IRAP, and LRP1 as well as several auxiliary components. Major IRV proteins have relatively long half-life inside the cell and survive multiple rounds of translocation to and from the cell surface. Here, we summarize evidence showing how the IRVs are self-assembled from pre-synthesized Glut4, sortilin, IRAP, and LRP1 after each translocation event. Furthermore, the cytoplasmic tail of sortilin binds Akt while cytoplasmic tails of IRAP and LRP1 interact with the Akt target, TBC1D4. Recruitment of signaling proteins to the IRVs may render insulin responsiveness to this compartment and thus distinguish it from other intracellular membrane vesicles.
Collapse
Affiliation(s)
- Konstantin V Kandror
- Department of Biochemistry and Cell Biology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States.
| |
Collapse
|
32
|
Zhou B, Ling L, Wang B, Yang F, Hou M, Liu F, Li Y, Luo H, He W, Ye H. Hepatopancreas Transcriptome Analysis of Spinibarbus sinensis to Reveal Different Growth-Related Genes. Genes (Basel) 2024; 15:949. [PMID: 39062728 PMCID: PMC11276559 DOI: 10.3390/genes15070949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Spinibarbus sinensis, also known as Qingbo, is an important economic fish in China. However, the detailed mechanisms underlying its growth are still unknown. To excavate the genes and signaling pathways related to its growth, we compared the transcriptome profiles of the hepatopancreas tissues of S. sinensis, with two groups of growth rate for evaluation. An average of 66,304,909 and 68,739,585 clean reads were obtained in the fast growth (FG) and slow growth (SG) group, respectively. The differential gene expression analysis results showed that 272 differentially expressed genes (DEGs) were screened between the FG and SG groups, including 101 up-regulated genes and 171 down-regulated genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis results showed that GO terms related to metabolic process, organic substance metabolic process, and catalytic activity were enriched, pathway signals related to steroid biosynthesis and protein digestion and absorption were also detected. Meanwhile, the potential key regulatory genes sst2, fndc4, and cckra related to the growth of S. sinensis were screened. Reverse transcript fluorescence quantitative PCR (RT-qPCR) validation of 18 DEGs associated with growth differences showed that the RT-qPCR results were consistent with RNA-seq analysis, and nine genes, stk31, gpr149, angptl1, fstl1, sik1, ror2, nlrc3, pdlim2, and nav2 were significantly expressed in the FG group. bmp1, stc1, gpatch8, sstrt2, s100a1, ktf6, cckar6, sync1, bhlha15, a total of nine genes were significantly expressed in the SG group. This study provides basic information for improving the growth characteristics of S. sinensis and the functional research of candidate genes.
Collapse
Affiliation(s)
- Bo Zhou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Yibin 644000, China; (B.Z.); (B.W.); (F.Y.)
| | - Leyan Ling
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| | - Bin Wang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Yibin 644000, China; (B.Z.); (B.W.); (F.Y.)
| | - Fei Yang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Yibin 644000, China; (B.Z.); (B.W.); (F.Y.)
| | - Mengdan Hou
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| | - Fan Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| | - Yu Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| | - Hui Luo
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| | - Wenping He
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| | - Hua Ye
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, College of Fisheries, Southwest University, Chongqing 402460, China; (L.L.); (M.H.); (F.L.); (Y.L.); (H.L.); (W.H.)
| |
Collapse
|
33
|
Huang X, Liang W, Yang R, Jin L, Zhao K, Chen J, Shang X, Zhou Y, Wang X, Yu H. Variations in the LINGO2 and GLIS3 Genes and Gene-Environment Interactions Increase Gestational Diabetes Mellitus Risk in Chinese Women. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:11596-11605. [PMID: 38888423 DOI: 10.1021/acs.est.4c03221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Gestational diabetes mellitus (GDM) has been found to be a common complication in pregnant women, known to escalate the risk of negative obstetric outcomes. In our study, we genotyped 1,566 Chinese pregnant women for two single nucleotide polymorphisms (SNPs) in the LINGO2 gene and one SNP in the GLIS3 gene, utilizing targeted next-generation sequencing. The impact of two interacting genes, and the interaction of genes with the environment─including exposure to particulate matter (PM2.5), ozone (O3), and variations in prepregnancy body mass index (BMI)─on the incidence of GDM were analyzed using logistic regression. Our findings identify the variants LINGO2 rs10968576 (P = 0.022, OR = 1.224) and rs1412239 (P = 0.018, OR = 1.231), as well as GLIS3 rs10814916 (P = 0.028, OR = 1.172), as risk mutations significantly linked to increased susceptibility to GDM. Further analysis underscores the crucial role of gene-gene and gene-environment interactions in the development of GDM among Chinese women (P < 0.05). Particularly, the individuals carrying the rs10968576 G-rs1412239 G-rs10814916 C haplotype exhibit increased susceptibility to GDM during the prepregnancy period when interacting with PM2.5, O3, and BMI (P = 8.004 × 10-7, OR = 1.206; P = 6.3264 × 10-11, OR = 1.280; P = 9.928 × 10-7, OR = 1.334, respectively). In conclusion, our research emphasizes the importance of the interaction between specific gene variations─LINGO2 and GLIS3─and environmental factors in influencing GDM risk. Notably, we found significant associations between these gene variations and GDM risk across various environmental exposure periods.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Weiwei Liang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Runqiu Yang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Lei Jin
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University, Beijing 100091, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Juan Chen
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xuejun Shang
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210023, China
| | - Yuanzhong Zhou
- School of Public Health, Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi 563000, China
| | - Xin Wang
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
34
|
Chiang WY, Yu HW, Wu MC, Huang YM, Chen YQ, Lin JW, Liu YW, You LR, Chiou A, Kuo JC. Matrix mechanics regulates muscle regeneration by modulating kinesin-1 activity. Biomaterials 2024; 308:122551. [PMID: 38593710 DOI: 10.1016/j.biomaterials.2024.122551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Sarcopenia, a prevalent muscle disease characterized by muscle mass and strength reduction, is associated with impaired skeletal muscle regeneration. However, the influence of the biomechanical properties of sarcopenic skeletal muscle on the efficiency of the myogenic program remains unclear. Herein, we established a mouse model of sarcopenia and observed a reduction in stiffness within the sarcopenic skeletal muscle in vivo. To investigate whether the biomechanical properties of skeletal muscle directly impact the myogenic program, we established an in vitro system to explore the intrinsic mechanism involving matrix stiffness control of myogenic differentiation. Our findings identify the microtubule motor protein, kinesin-1, as a mechano-transduction hub that senses and responds to matrix stiffness, crucial for myogenic differentiation and muscle regeneration. Specifically, kinesin-1 activity is positively regulated by stiff matrices, facilitating its role in transporting mitochondria and enhancing translocation of the glucose transporter GLUT4 to the cell surface for glucose uptake. Conversely, the softer matrices significantly suppress kinesin-1 activity, leading to the accumulation of mitochondria around nuclei and hindering glucose uptake by inhibiting GLUT4 membrane translocation, consequently impairing myogenic differentiation. The insights gained from the in-vitro system highlight the mechano-transduction significance of kinesin-1 motor proteins in myogenic differentiation. Furthermore, our study confirms that enhancing kinesin-1 activity in the sarcopenic mouse model restores satellite cell expansion, myogenic differentiation, and muscle regeneration. Taken together, our findings provide a potential target for improving muscle regeneration in sarcopenia.
Collapse
Affiliation(s)
- Wan-Yu Chiang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yi-Man Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yin-Quan Chen
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Jong-Wei Lin
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yen-Wenn Liu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Li-Ru You
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan; Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
| |
Collapse
|
35
|
Park SH, Lee H. Diabetes Mellitus Inequality in South Korean Adults by Region: The Influence of Obesity and Depression. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:1508-1516. [PMID: 39086420 PMCID: PMC11287605 DOI: 10.18502/ijph.v53i7.16045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/10/2023] [Indexed: 08/02/2024]
Abstract
Background Diabetes mellitus (DM) has different prevalence by region. This study aimed to identify the differences in the effects of obesity and depression on DM in South Korean adults by region. Methods The participants were 14,343 adults (≥30 yr) from Ulsan (regions with the lowest prevalence of DM) and Jeonbuk (regions with the highest prevalence of DM), and data were extracted from the Community Health Survey 2019. We applied a complex sampling design analysis to reflect the stratified, clustering and weights. The data were analyzed using the unweighted frequencies, weighted percentage, mean, standard error, Chi-Square test and multiple logistic regression analysis (SPSS 25.0). Results Regarding the main result for Ulsan, the odds ratio of DM increased by 1.94, 2.52,1.57, and 4.87 times for obesity(25-29.9kg/m2), high obesity(≥30kg/m2), depression, and receipt of psychological counseling for depression, respectively. In Jeonbuk, the odds ratio of DM increased by 1.79, 2.84, and 3.59 times for obesity, high obesity, and unmet medical experience, respectively. On the other hand, depression-related variables were found to not influence DM. Conclusion We provided the rationale for conducting a health project that interventions for obesity and depression should be included in DM management programs differently in Ulsan and Jeonbuk regions.
Collapse
Affiliation(s)
- Seong-Hi Park
- School of Nursing, Soonchunhyang University, Asan, South Korea
| | - Heashoon Lee
- School of Nursing, Hannam University, Daejeon, South Korea
| |
Collapse
|
36
|
Kołodziej-Sobczak D, Sobczak Ł, Łączkowski KZ. Protein Tyrosine Phosphatase 1B (PTP1B): A Comprehensive Review of Its Role in Pathogenesis of Human Diseases. Int J Mol Sci 2024; 25:7033. [PMID: 39000142 PMCID: PMC11241624 DOI: 10.3390/ijms25137033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Overexpression of protein tyrosine phosphatase 1B (PTP1B) disrupts signaling pathways and results in numerous human diseases. In particular, its involvement has been well documented in the pathogenesis of metabolic disorders (diabetes mellitus type I and type II, fatty liver disease, and obesity); neurodegenerative diseases (Alzheimer's disease, Parkinson's disease); major depressive disorder; calcific aortic valve disease; as well as several cancer types. Given this multitude of therapeutic applications, shortly after identification of PTP1B and its role, the pursuit to introduce safe and selective enzyme inhibitors began. Regrettably, efforts undertaken so far have proved unsuccessful, since all proposed PTP1B inhibitors failed, or are yet to complete, clinical trials. Intending to aid introduction of the new generation of PTP1B inhibitors, this work collects and organizes the current state of the art. In particular, this review intends to elucidate intricate relations between numerous diseases associated with the overexpression of PTP1B, as we believe that it is of the utmost significance to establish and follow a brand-new holistic approach in the treatment of interconnected conditions. With this in mind, this comprehensive review aims to validate the PTP1B enzyme as a promising molecular target, and to reinforce future research in this direction.
Collapse
Affiliation(s)
- Dominika Kołodziej-Sobczak
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089 Bydgoszcz, Poland;
| | - Łukasz Sobczak
- Hospital Pharmacy, Multidisciplinary Municipal Hospital in Bydgoszcz, Szpitalna 19, 85-826 Bydgoszcz, Poland
| | - Krzysztof Z. Łączkowski
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089 Bydgoszcz, Poland;
| |
Collapse
|
37
|
Das M, Chakraborty M, Das P, Santra S, Mukherjee A, Das S, Banyai K, Roy S, Choudhury L, Gupta R, Dey T, Das D, Bose A, Ganesh B, Banerjee R. System biology approaches for systemic diseases: Emphasis on type II diabetes mellitus and allied metabolism. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2024; 58:103176. [DOI: 10.1016/j.bcab.2024.103176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
38
|
Zhang T, Yi Q, Huang W, Feng J, Liu H. New insights into the roles of Irisin in diabetic cardiomyopathy and vascular diseases. Biomed Pharmacother 2024; 175:116631. [PMID: 38663105 DOI: 10.1016/j.biopha.2024.116631] [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: 02/05/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 06/03/2024] Open
Abstract
Diabetes mellitus (DM) is a prevalent chronic disease in the 21st century due to increased lifespan and unhealthy lifestyle choices. Extensive research indicates that exercise can play a significant role in regulating systemic metabolism by improving energy metabolism and mitigating various metabolic disorders, including DM. Irisin, a well-known exerkine, was initially reported to enhance energy expenditure by indicating the browning of white adipose tissue (WAT) through peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) signaling. In this review, we summarize the potential mechanisms underlying the beneficial effects of Irisin on glucose dysmetabolism, including reducing gluconeogenesis, enhancing insulin energy expenditure, and promoting glycogenesis. Additionally, we highlight Irisin's potential to improve diabetic vascular diseases by stimulating nitric oxide (NO) production, reducing oxidative and nitrosative stress, curbing inflammation, and attenuating endothelial cell aging. Furthermore, we discuss the potential of Irisin to improve diabetic cardiomyopathy by preventing cardiomyocyte loss and reducing myocardial hypertrophy and fibrosis. Given Irisin's promising functions in managing diabetic cardiomyopathy and vascular diseases, targeting Irisin for therapeutic purposes could be a fruitful avenue for future research and clinical interventions.
Collapse
Affiliation(s)
- Tiandong Zhang
- Collage of Integration of Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qian Yi
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Wenhua Huang
- Collage of Integration of Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China; Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Jianguo Feng
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; The Third People's Hospital of Longmatan District, Luzhou, Sichuan 646000, China.
| |
Collapse
|
39
|
Araujo L, Dias C, Sucupira F, Ramalho L, Camporez J. A short-term rodent model for non-alcoholic steatohepatitis induced by a high-fat diet and carbon tetrachloride. Biosci Rep 2024; 44:BSR20231532. [PMID: 38660995 PMCID: PMC11081943 DOI: 10.1042/bsr20231532] [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: 10/06/2023] [Revised: 02/15/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
Several models of mice-fed high-fat diets have been used to trigger non-alcoholic steatohepatitis and some chemical substances, such as carbon tetrachloride. The present study aimed to evaluate the joint action of a high-fat diet and CCl4 in developing a short-term non-alcoholic steatohepatitis model. C57BL6/J mice were divided into two groups: standard diet-fed (SD), the high-fat diet-fed (HFD) and HFD + fructose-fed and carbon tetrachloride (HFD+CCl4). The animals fed with HFD+CCl4 presented increased lipid deposition compared with both SD and HFD mice. Plasma cholesterol was increased in animals from the HFD+CCl4 group compared with the SD and HFD groups, without significant differences between the SD and HFD groups. Plasma triglycerides showed no significant difference between the groups. The HFD+CCl4 animals had increased collagen deposition in the liver compared with both SD and HFD groups. Hydroxyproline was also increased in the HFD+CCl4 group. Liver enzymes, alanine aminotransferase and aspartate aminotransferase, were increased in the HFD+CCl4 group, compared with SD and HFD groups. Also, CCl4 was able to trigger an inflammatory process in the liver of HFD-fed animals by promoting an increase of ∼2 times in macrophage activity, ∼6 times in F4/80 gene expression, and pro-inflammatory cytokines (IL-1b and TNFa), in addition to an increase in inflammatory pathway protein phosphorylation (IKKbp). HFD e HFD+CCl4 animals increased glucose intolerance compared with SD mice, associated with reduced insulin-stimulated AKT activity in the liver. Therefore, our study has shown that short-term HFD feeding associated with fructose and CCl4 can trigger non-alcoholic steatohepatitis and cause damage to glucose metabolism.
Collapse
Affiliation(s)
- Layanne C.C. Araujo
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Brazil
| | - Carolina C.B. Dias
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Brazil
| | - Felipe G. Sucupira
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Brazil
| | - Leandra N.Z. Ramalho
- Department of Pathology and Legal Medicine, Ribeirao Preto School of Medicine, University of Sao Paulo, Brazil
| | - João Paulo Camporez
- Department of Physiology, Ribeirao Preto School of Medicine, University of Sao Paulo, Brazil
| |
Collapse
|
40
|
Zhao Z, Xiang X, Chen Q, Du J, Zhu S, Xu X, Shen Y, Wen S, Li Y, Xu W, Mai K, Ai Q. Sterol Regulatory Element Binding Protein 1: A Mediator for High-Fat Diet-Induced Hepatic Gluconeogenesis and Glucose Intolerance in Fish. J Nutr 2024; 154:1505-1516. [PMID: 38460786 DOI: 10.1016/j.tjnut.2024.02.031] [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: 01/23/2024] [Accepted: 02/26/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Sterol regulatory element binding protein (SREBP) 1 is considered to be a crucial regulator for lipid synthesis in vertebrates. However, whether SREBP1 could regulate hepatic gluconeogenesis under high-fat diet (HFD) condition is still unknown, and the underlying mechanism is also unclear. OBJECTIVES This study aimed to determine gluconeogenesis-related gene and protein expressions in response to HFD in large yellow croaker and explore the role and mechanism of SREBP1 in regulating the related transcription and signaling. METHODS Croakers (mean weight, 15.61 ± 0.10 g) were fed with diets containing 12% crude lipid [control diet (ND)] or 18% crude lipid (HFD) for 10 weeks. The glucose tolerance, insulin tolerance, hepatic gluconeogenesis-related genes, and proteins expressions were determined. To explore the role of SREBP1 in HFD-induced gluconeogenesis, SREBP1 was inhibited by pharmacologic inhibitor (fatostatin) or genetic knockdown in croaker hepatocytes under palmitic acid (PA) condition. To explore the underlying mechanism, luciferase reporter and chromatin immunoprecipitation assays were conducted in HEK293T cells. Data were analyzed using analysis of variance or Student t test. RESULTS Compared with ND, HFD increased the mRNA expressions of gluconeogenesis genes (2.40-fold to 2.60-fold) (P < 0.05) and reduced protein kinase B (AKT) phosphorylation levels (0.28-fold to 0.34-fold) (P < 0.05) in croakers. However, inhibition of SREBP1 by fatostatin addition or SREBP1 knockdown reduced the mRNA expressions of gluconeogenesis genes (P < 0.05) and increased AKT phosphorylation levels (P < 0.05) in hepatocytes, compared with that by PA treatment. Moreover, fatostatin addition or SREBP1 knockdown also increased the mRNA expressions of irs1 (P < 0.05) and reduced serine phosphorylation of IRS1 (P < 0.05). Furthermore, SREBP1 inhibited IRS1 transcriptions by binding to its promoter and induced IRS1 serine phosphorylation by activating diacylglycerol-protein kinase Cε signaling. CONCLUSIONS This study reveals the role of SREBP1 in hepatic gluconeogenesis under HFD condition in croakers, which may provide a potential strategy for improving HFD-induced glucose intolerance.
Collapse
Affiliation(s)
- Zengqi Zhao
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Xiaojun Xiang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Qiang Chen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Jianlong Du
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Si Zhu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Xiang Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Yanan Shen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Shunlang Wen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Yueru Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Wei Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, Shandong, China.
| |
Collapse
|
41
|
Priscilla L, Yoo C, Jang S, Park S, Lim G, Kim T, Lee DY. Immunotherapy targeting the obese white adipose tissue microenvironment: Focus on non-communicable diseases. Bioact Mater 2024; 35:461-476. [PMID: 38404641 PMCID: PMC10884763 DOI: 10.1016/j.bioactmat.2024.01.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/14/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Obesity triggers inflammatory responses in the microenvironment of white adipose tissue, resulting in chronic systemic inflammation and the subsequent development of non-communicable diseases, including type 2 diabetes, coronary heart disease, and breast cancer. Current therapy approaches for obesity-induced non-communicable diseases persist in prioritizing symptom remission while frequently overlooking the criticality of targeting and alleviating inflammation at its source. Accordingly, this review highlights the importance of the microenvironment of obese white adipose tissue and the promising potential of employing immunotherapy to target it as an effective therapeutic approach for non-communicable diseases induced by obesity. Additionally, this review discusses the challenges and offers perspective about the immunotherapy targeting the microenvironment of obese white adipose tissue.
Collapse
Affiliation(s)
- Lia Priscilla
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Chaerim Yoo
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Seonmi Jang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Sewon Park
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Gayoung Lim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Taekyun Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Nano Science and Technology (INST) & Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul, 04763, Republic of Korea
- Elixir Pharmatech Inc., Seoul, 07463, Republic of Korea
| |
Collapse
|
42
|
Kim MJ, Heo M, Kim SJ, Song HE, Lee H, Kim NE, Shin H, Do AR, Kim J, Cho YM, Hong YS, Kim WJ, Won S, Yoo HJ. Associations between plasma metabolites and heavy metal exposure in residents of environmentally polluted areas. ENVIRONMENT INTERNATIONAL 2024; 187:108709. [PMID: 38723457 DOI: 10.1016/j.envint.2024.108709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/08/2024] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Heavy metals are commonly released into the environment through industrial processes such as mining and refining. The rapid industrialization that occurred in South Korea during the 1960s and 1970s contributed significantly to the economy of the country; however, the associated mining and refining led to considerable environmental pollution, and although mining is now in decline in South Korea, the detrimental effects on residents inhabiting the surrounding areas remain. The bioaccumulation of toxic heavy metals leads to metabolic alterations in human homeostasis, with disruptions in this balance leading to various health issues. This study used metabolomics to explore metabolomic alterations in the plasma samples of residents living in mining and refining areas. The results showed significant increases in metabolites involved in glycolysis and the surrounding metabolic pathways, such as glucose-6-phosphate, phosphoenolpyruvate, lactate, and inosine monophosphate, in those inhabiting polluted areas. An investigation of the associations between metabolites and blood clinical parameters through meet-in-the-middle analysis indicated that female residents were more affected by heavy metal exposure, resulting in more metabolomic alterations. For women, inhabiting the abandoned mine area, metabolites in the glycolysis and pentose phosphate pathways, such as ribose-5-phosphate and 3-phosphoglycerate, have shown a negative correlation with albumin and calcium. Finally, Mendelian randomization(MR) was used to determine the causal effects of these heavy metal exposure-related metabolites on heavy metal exposure-related clinical parameters. Metabolite biomarkers could provide insights into altered metabolic pathways related to exposure to toxic heavy metals and improve our understanding of the molecular mechanisms underlying the health effects of toxic heavy metal exposure.
Collapse
Affiliation(s)
- Mi Jeong Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Min Heo
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Su Jung Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Ha Eun Song
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Hyoyeong Lee
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Nam-Eun Kim
- Department of Public Health Sciences, Seoul National University, Seoul, South Korea
| | - Hyeongyu Shin
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Ah Ra Do
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, South Korea; RexSoft Corp, Seoul, South Korea
| | - Jeeyoung Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Yong Min Cho
- Department of Nano Chemical and Biological Engineering, Seokyeong University, Seoul, Republic of Korea
| | - Young-Seoub Hong
- Department of Preventive Medicine, College of Medicine, Dong-A University, 32, Daesin Gongwon-ro, Seo-gu, Busan 49201, Korea
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Sungho Won
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, South Korea; Department of Public Health Sciences, Seoul National University, Seoul, South Korea; Institute of Health and Environment, Seoul National University, Seoul, South Korea; RexSoft Corp, Seoul, South Korea.
| | - Hyun Ju Yoo
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea; Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea; Department of Digital Medicine, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
43
|
Wang Y. Fasting Triglycerides in the Upper Normal Range Are Independently Associated with an Increased Risk of Diabetes Mortality in a Large Representative US Population. J Cardiovasc Dev Dis 2024; 11:128. [PMID: 38667746 PMCID: PMC11050947 DOI: 10.3390/jcdd11040128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
The association between normal-range triglyceride levels and diabetes mortality remains unclear. This cohort study aimed to elucidate this relationship by examining 19,010 US adult participants with fasting serum triglycerides below 150 mg/dL. Cox proportional hazards models were employed to estimate mortality hazard ratios (HRs) and 95% confidence intervals (CIs). Participants were followed up for a mean of 15.3 years, during which 342 diabetes deaths were recorded. A 1 natural log unit increase in triglycerides was associated with a 57% higher risk of diabetes mortality (adjusted HR, 1.57; 95% CI, 1.04-2.38). Comparable results were obtained when triglycerides were analyzed in quartiles. Receiver operating characteristic curve analysis identified an optimal triglyceride cutoff of 94.5 mg/dL for diabetes mortality; individuals with triglyceride levels above this threshold faced a greater risk of diabetes mortality (adjusted HR, 1.43; 95% CI, 1.12-1.83). Further investigation revealed a positive association between normal triglyceride levels and all-cause mortality, though no association was observed between normal triglycerides and mortality from hypertension or cardiovascular disease. In conclusion, elevated triglyceride levels within the normal range were associated with an increased risk of diabetes mortality. Individuals with triglyceride levels of 95 mg/dL or higher may require vigilant monitoring for diabetes and its associated complications.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| |
Collapse
|
44
|
Rugg C, Schmid S, Zipperle J, Kreutziger J. Stress hyperglycaemia following trauma - a survival benefit or an outcome detriment? Curr Opin Anaesthesiol 2024; 37:131-138. [PMID: 38390910 DOI: 10.1097/aco.0000000000001350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
PURPOSE OF REVIEW Stress hyperglycaemia occur often in critically injured patients. To gain new consideration about it, this review compile current as well as known immunological and biochemical findings about causes and emergence. RECENT FINDINGS Glucose is the preferred energy substrate for fending immune cells, reparative tissue and the cardiovascular system following trauma. To fulfil these energy needs, the liver is metabolically reprogrammed to rebuild glucose from lactate and glucogenic amino acids (hepatic insulin resistance) at the expenses of muscles mass and - to a less extent - fat tissue (proteolysis, lipolysis, peripheral insulin resistance). This inevitably leads to stress hyperglycaemia, which is evolutionary preserved and seems to be an essential and beneficial survival response. It is initiated by damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), intensified by immune cells itself and mainly ruled by tumour necrosis factor (TNF)α and catecholamines with lactate and hypoxia inducible factor (HIF)-1α as intracellular signals and lactate as an energy shuttle. Important biochemical mechanisms involved in this response are the Warburg effect as an efficient metabolic shortcut and the extended Cori cycle. SUMMARY Stress hyperglycaemia is beneficial in an acute life-threatening situation, but further research is necessary, to prevent trauma patients from the detrimental effects of persisting hyperglycaemia.
Collapse
Affiliation(s)
- Christopher Rugg
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schmid
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Zipperle
- Johannes Zipperle, Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Janett Kreutziger
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
45
|
Glatz JFC, Heather LC, Luiken JJFP. CD36 as a gatekeeper of myocardial lipid metabolism and therapeutic target for metabolic disease. Physiol Rev 2024; 104:727-764. [PMID: 37882731 DOI: 10.1152/physrev.00011.2023] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023] Open
Abstract
The multifunctional membrane glycoprotein CD36 is expressed in different types of cells and plays a key regulatory role in cellular lipid metabolism, especially in cardiac muscle. CD36 facilitates the cellular uptake of long-chain fatty acids, mediates lipid signaling, and regulates storage and oxidation of lipids in various tissues with active lipid metabolism. CD36 deficiency leads to marked impairments in peripheral lipid metabolism, which consequently impact on the cellular utilization of multiple different fuels because of the integrated nature of metabolism. The functional presence of CD36 at the plasma membrane is regulated by its reversible subcellular recycling from and to endosomes and is under the control of mechanical, hormonal, and nutritional factors. Aberrations in this dynamic role of CD36 are causally associated with various metabolic diseases, in particular insulin resistance, diabetic cardiomyopathy, and cardiac hypertrophy. Recent research in cardiac muscle has disclosed the endosomal proton pump vacuolar-type H+-ATPase (v-ATPase) as a key enzyme regulating subcellular CD36 recycling and being the site of interaction between various substrates to determine cellular substrate preference. In addition, evidence is accumulating that interventions targeting CD36 directly or modulating its subcellular recycling are effective for the treatment of metabolic diseases. In conclusion, subcellular CD36 localization is the major adaptive regulator of cellular uptake and metabolism of long-chain fatty acids and appears a suitable target for metabolic modulation therapy to mend failing hearts.
Collapse
Affiliation(s)
- Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lisa C Heather
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
46
|
Wang Y, Fang Y, Vrablik M. Homeostasis Model Assessment for Insulin Resistance Mediates the Positive Association of Triglycerides with Diabetes. Diagnostics (Basel) 2024; 14:733. [PMID: 38611646 PMCID: PMC11011406 DOI: 10.3390/diagnostics14070733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Elevated circulating triglyceride levels have been linked to an increased risk of diabetes, although the precise mechanisms remain unclear. This study aimed to investigate whether low-density lipoprotein (LDL) cholesterol, homeostatic model assessment (HOMA) for insulin resistance, and C-reactive protein (CRP) served as mediators in this association across a sample of 18,435 US adults. Mediation analysis was conducted using the PROCESS Version 4.3 Macro for SPSS. Simple mediation analysis revealed that all three potential mediators played a role in mediating the association. However, in parallel mediation analysis, where all three mediators were simultaneously included, HOMA for insulin resistance remained a significant mediator (indirect effect coefficient, 0.47; 95% confidence interval [CI], 0.43-0.52; p < 0.05) after adjusting for all tested confounding factors. Conversely, LDL cholesterol (indirect effect coefficient, -0.13; 95% CI, -0.31-0.05; p > 0.05) and C-reactive protein (indirect effect coefficient, 0.01; 95% CI, -0.003-0.02; p > 0.05) ceased to be significant mediators. HOMA for insulin resistance accounted for 49% of the association between triglycerides and diabetes. In conclusion, HOMA for insulin resistance was the dominant mediator underlying the association between triglycerides and diabetes. Therefore, reducing triglyceride levels may hold promise for improving insulin sensitivity in diabetic patients.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Yan Fang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Michal Vrablik
- Third Department of Medicine, General University Hospital and First Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic;
| |
Collapse
|
47
|
Slusher AL, Nouws J, Tokoglu F, Vash-Margita A, Matthews MD, Fitch M, Shankaran M, Hellerstein MK, Caprio S. Altered extracellular matrix dynamics is associated with insulin resistance in adolescent children with obesity. Obesity (Silver Spring) 2024; 32:593-602. [PMID: 38410080 PMCID: PMC11034857 DOI: 10.1002/oby.23974] [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: 10/11/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 02/28/2024]
Abstract
OBJECTIVE The objective of this study was to examine the hypothesis that abdominal and gluteal adipocyte turnover, lipid dynamics, and fibrogenesis are dysregulated among insulin-resistant (IR) compared with insulin-sensitive (IS) adolescents with obesity. METHODS Seven IS and seven IR adolescents with obesity participated in a 3-h oral glucose tolerance test and a multi-section magnetic resonance imaging scan of the abdominal region to examine body fat distribution patterns and liver fat content. An 8-week 70% deuterated water (2 H2 O) labeling protocol examined adipocyte turnover, lipid dynamics, and fibrogenesis in vivo from biopsied abdominal and gluteal fat. RESULTS Abdominal and gluteal subcutaneous adipose tissue (SAT) turnover rates of lipid components were similar among IS and IR adolescents with obesity. However, the insoluble collagen (type I, subunit α2) isoform measured from abdominal, but not gluteal, SAT was elevated in IR compared with IS individuals. In addition, abdominal insoluble collagen Iα2 was associated with ratios of visceral-to-total (visceral adipose tissue + SAT) abdominal fat and whole-body and adipose tissue insulin signaling, and it trended toward a positive association with liver fat content. CONCLUSIONS Altered extracellular matrix dynamics, but not expandability, potentially decreases abdominal SAT lipid storage capacity, contributing to the pathophysiological pathways linking adipose tissue and whole-body IR with altered ectopic storage of lipids within the liver among IR adolescents with obesity.
Collapse
Affiliation(s)
- Aaron L. Slusher
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Jessica Nouws
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Fuyuze Tokoglu
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Alla Vash-Margita
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Marcy D. Matthews
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Mark Fitch
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Sonia Caprio
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
48
|
Greeny A, Nair A, Sadanandan P, Satarker S, Famurewa AC, Nampoothiri M. Epigenetic Alterations in Alzheimer's Disease: Impact on Insulin Signaling and Advanced Drug Delivery Systems. BIOLOGY 2024; 13:157. [PMID: 38534427 DOI: 10.3390/biology13030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that predominantly affects the hippocampus and the entorhinal complex, leading to memory lapse and cognitive impairment. This can have a negative impact on an individual's behavior, speech, and ability to navigate their surroundings. AD is one of the principal causes of dementia. One of the most accepted theories in AD, the amyloid β (Aβ) hypothesis, assumes that the buildup of the peptide Aβ is the root cause of AD. Impaired insulin signaling in the periphery and central nervous system has been considered to have an effect on the pathophysiology of AD. Further, researchers have shifted their focus to epigenetic mechanisms that are responsible for dysregulating major biochemical pathways and intracellular signaling processes responsible for directly or indirectly causing AD. The prime epigenetic mechanisms encompass DNA methylation, histone modifications, and non-coding RNA, and are majorly responsible for impairing insulin signaling both centrally and peripherally, thus leading to AD. In this review, we provide insights into the major epigenetic mechanisms involved in causing AD, such as DNA methylation and histone deacetylation. We decipher how the mechanisms alter peripheral insulin signaling and brain insulin signaling, leading to AD pathophysiology. In addition, this review also discusses the need for newer drug delivery systems for the targeted delivery of epigenetic drugs and explores targeted drug delivery systems such as nanoparticles, vesicular systems, networks, and other nano formulations in AD. Further, this review also sheds light on the future approaches used for epigenetic drug delivery.
Collapse
Affiliation(s)
- Alosh Greeny
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ayushi Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Prashant Sadanandan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo 482123, Nigeria
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
49
|
Waritu NC, Nair SKP, Birhan B, Adugna T, Awgichew GB, Jemal M. Serum Lipid Profiles, Blood Glucose, and High-Sensitivity C-Reactive Protein Levels Among People Living with HIV Taking Dolutegravir and Ritonavir-Boosted Atazanavir-Based Antiretroviral Therapy at Jimma University Medical Center, Southwest Ethiopia, 2021. HIV AIDS (Auckl) 2024; 16:17-32. [PMID: 38369986 PMCID: PMC10873129 DOI: 10.2147/hiv.s430310] [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: 09/29/2023] [Accepted: 02/05/2024] [Indexed: 02/20/2024] Open
Abstract
Background Long-term use of antiretroviral therapy, especially dolutegravir and boosted-atazanavir, raises concerns about cardiovascular disease. Thus, this study aimed to assess lipid profiles, blood glucose, and high-sensitivity C-reactive protein levels among people living with HIV on dolutegravir and ritonavir-boosted atazanavir-based therapy. Methods An institutional-based comparative cross-sectional study was conducted from November 4, 2021, to January 4, 2022. An equal number of dolutegravir- and ritonavir-boosted atazanavir-treated patients (n = 64 each) was enrolled. A consecutive sampling was used to select participants. The Chi-square, Student's t-test, Mann-Whitney U-test, and logistic regression were used as appropriate statistical tests using SPSS Version 25.0. Statistical significance was set at p < 0.05. Results Dyslipidemia was found in 67.2% (43/64) of ritonavir-boosted atazanavir group and 48.4% (31/64) of dolutegravir group. The dolutegravir group had significantly higher mean and median values of high-density lipoprotein and random blood sugar, respectively, as well as lower median triglyceride and high-sensitivity C-reactive protein levels than the ritonavir-boosted atazanavir group. Ritonavir-boosted atazanavir-based regimens (AOR=3.4, 95% CI: 1.5, 8) and age >40 years were predictors of dyslipidemia, while BMI ≥25 kg/m2 (AOR=3.7, 95% CI: 1.3, 10.8) and dolutegravir-based regimens (AOR=4.6, 95% CI: 1.5, 14) were predictors of hyperglycemia. Ritonavir-boosted atazanavir-based regimens (ARR=3, 95% CI: 1.3, 8) and BMI ≥25 kg/m2 (ARR=2.5, 95% CI: 1.1, 6) were associated with increased high-sensitivity C-reactive protein by 1-3 mg/L. The risk of increased high-sensitivity C-reactive protein by >3 mg/L was greater in those patients with a CD4 cell count of <500 cells/mm3 (ARR=5, 95% CI: 1.1, 24). Conclusion When compared to ritonavir-boosted atazanavir-based regimens, dolutegravir had favorable lipid profiles and high-sensitivity C-reactive protein but unfavorable blood glucose levels. Therefore, baseline blood glucose, lipid profiles, and high-sensitivity C-reactive protein levels should be routinely measured in patients on these regimens.
Collapse
Affiliation(s)
- Nuredin Chura Waritu
- Department of Biomedical Sciences, School of Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Suresh Kumar P Nair
- Department of Biomedical Sciences, School of Medicine, Jimma University, Jimma, Ethiopia
| | - Bihonegn Birhan
- Department of Biomedical Sciences, School of Medicine, Jimma University, Jimma, Ethiopia
| | - Tesfaye Adugna
- Department of Biomedical Sciences, School of Medicine, Jimma University, Jimma, Ethiopia
| | - Gesese Bogale Awgichew
- Department of Biomedical Sciences, School of Medicine, Jimma University, Jimma, Ethiopia
| | - Mohammed Jemal
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
50
|
Chandrasekaran P, Weiskirchen R. The Role of Obesity in Type 2 Diabetes Mellitus-An Overview. Int J Mol Sci 2024; 25:1882. [PMID: 38339160 PMCID: PMC10855901 DOI: 10.3390/ijms25031882] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity or excessive weight gain is identified as the most important and significant risk factor in the development and progression of type 2 diabetes mellitus (DM) in all age groups. It has reached pandemic dimensions, making the treatment of obesity crucial in the prevention and management of type 2 DM worldwide. Multiple clinical studies have demonstrated that moderate and sustained weight loss can improve blood glucose levels, insulin action and reduce the need for diabetic medications. A combined approach of diet, exercise and lifestyle modifications can successfully reduce obesity and subsequently ameliorate the ill effects and deadly complications of DM. This approach also helps largely in the prevention, control and remission of DM. Obesity and DM are chronic diseases that are increasing globally, requiring new approaches to manage and prevent diabetes in obese individuals. Therefore, it is essential to understand the mechanistic link between the two and design a comprehensive approach to increase life expectancy and improve the quality of life in patients with type 2 DM and obesity. This literature review provides explicit information on the clinical definitions of obesity and type 2 DM, the incidence and prevalence of type 2 DM in obese individuals, the indispensable role of obesity in the pathophysiology of type 2 DM and their mechanistic link. It also discusses clinical studies and outlines the recent management approaches for the treatment of these associated conditions. Additionally, in vivo studies on obesity and type 2 DM are discussed here as they pave the way for more rigorous development of therapeutic approaches.
Collapse
Affiliation(s)
- Preethi Chandrasekaran
- UT Southwestern Medical Center Dallas, 5323 Harry Hines Blvd. ND10.504, Dallas, TX 75390-9014, USA
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH), University Hospital Aachen, D-52074 Aachen, Germany
| |
Collapse
|