1
|
Kaylan KB, Nargis T, Figatner K, Wang JE, Pratuangtham S, Chakraborty A, Casimiro I, Nadler JL, Boxer MB, Maloney DJ, Anderson RM, Mirmira RG, Tersey SA. 12-Lipoxygenase Inhibition Improves Glycemia and Obesity-associated Inflammation in Male Human Gene Replacement Mice. Endocrinology 2025; 166:bqaf069. [PMID: 40186458 PMCID: PMC12022223 DOI: 10.1210/endocr/bqaf069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 04/07/2025]
Abstract
Obesity-associated inflammation is characterized by macrophage infiltration into peripheral tissues, contributing to the progression of prediabetes and type 2 diabetes. 12-lipoxygenase (12-LOX) catalyzes the formation of pro-inflammatory eicosanoids and promotes the migration of macrophages, yet its role in obesity-associated inflammation remains incompletely understood. Furthermore, differences between mouse and human orthologs of 12-LOX have limited efforts to study existing pharmacologic inhibitors of 12-LOX. In this study, we used a human gene replacement mouse model in which the gene encoding mouse 12-LOX (Alox15) is replaced by the human ALOX12 gene. As a model of obesity and dysglycemia, we administered male mice a high-fat diet. We subsequently investigated the effects of VLX-1005, a potent and selective small molecule inhibitor of human 12-LOX. Oral administration of VLX-1005 resulted in improved glucose homeostasis, decreased β-cell dedifferentiation, and reduced macrophage infiltration in islets and adipose tissue. Analysis of the stromal vascular fraction from adipose tissue showed a reduction in myeloid cells and cytokine expression with VLX-1005 treatment, indicating decreased adipose tissue inflammation. In a distinct mouse model in which Alox15 was selectively deleted in myeloid cells, we observed decreased β-cell dedifferentiation and reduced macrophage infiltration in both islets and adipose tissue, suggesting that the effects of VLX-1005 may relate to the inhibition of 12-LOX in macrophages. These findings highlight 12-LOX as a key factor in obesity-associated inflammation and suggest that 12-LOX inhibition could serve as a therapeutic strategy to improve glucose homeostasis and peripheral inflammation in the setting of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Kerim B Kaylan
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Titli Nargis
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Kayla Figatner
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jiayi E Wang
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarida Pratuangtham
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Advaita Chakraborty
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Isabel Casimiro
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jerry L Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | - Ryan M Anderson
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Raghavendra G Mirmira
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A Tersey
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
2
|
Su Y, Liu X, Jiang B, He H, Li F, Li X, Wang Y, Chen X, Wang X, Luo J, Chen L, Wu J, Hu F, Zhang M, Hu D, Ma J, Qin P. Potato Intake and the Risk of Overweight/Obesity, Hypertension, Diabetes, and Cardiovascular Disease: A Systematic Review and Meta-analysis of Observational Studies. Nutr Rev 2025; 83:466-478. [PMID: 39527022 DOI: 10.1093/nutrit/nuae159] [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] [Indexed: 11/16/2024] Open
Abstract
CONTEXT Results from observational studies and meta-analyses examining the relationship between total, fried, and nonfried potato intake and cardiometabolic disease remain conflicted. OBJECTIVE The aim was to synthesize existing evidence on the relationships between total potato intake and specific types of potato intake with the risk of overweight/obesity, hypertension, diabetes, gestational diabetes mellitus (GDM), cardiovascular disease (CVD), coronary heart disease (CHD), and stroke. DATA SOURCES Systematic searches were conducted in PubMed, Embase, and Web of Science until July 13, 2023. DATA EXTRACTION Data extracted from studies included first author, publication year, location, data source, follow-up duration (cohort studies only), demographics, sample size, cases, outcomes, exposure and outcome types, measurements, adjustments, study design, potato intake categories, and adjusted risk estimates (odds ratio, relative risk [RR], hazard ratio) with 95% CIs. DATA ANALYSIS Random-effects models were applied to estimate the summary RRs and 95% CIs. RESULTS Fifty-one articles (103 studies) were identified in the current meta-analysis. Comparing the highest with the lowest categories of total potato intake, total potato intake was not associated with hypertension (summary RR = 1.07; 95% CI: 0.95, 1.21), diabetes (1.08; 95% CI: 0.96, 1.22), GDM (1.16; 95% CI: 0.86, 1.57), CHD (1.00; 95% CI: 0.99, 1.02), CVD (0.97; 95% CI: 0.91, 1.03), or stroke (0.97; 95% CI: 0.88, 1.06); fried potato intake was not associated with overweight/obesity (1.24; 95% CI: 0.90, 1.70) or GDM (1.03; 95% CI: 0.97, 1.09) but was significantly associated with increased diabetes risk (1.16; 95% CI: 1.04, 1.30); nonfried potato intake was significantly associated with increased diabetes risk (1.05; 95% CI: 1.01, 1.10) but not hypertension (1.06; 95% CI: 0.97, 1.15). CONCLUSION Total potato intake was not associated with an increased risk of hypertension, diabetes, GDM, or cardiometabolic disease, but both fried and nonfried potato intake may increase the risk of diabetes but not other cardiometabolic diseases. Future cohort studies are needed to explore the association between different types of potato intake and cardiometabolic disease. In addition, the limited number of studies on total potato intake and overweight/obesity/heart failure, fried potato intake and CHD/stroke/heart failure, and nonfried potato intake and overweight/obesity/GDM/CHD/CVD/heart failure prevented us from conducting an analysis. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. RD42023454244.
Collapse
Affiliation(s)
- Yuhao Su
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
- School of Public Health, Shantou University, Shantou, Guangdong 515063, China
| | - Xiaoning Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Bin Jiang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Haitian He
- Urology Department, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Fengjuan Li
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
- School of Public Health, Shantou University, Shantou, Guangdong 515063, China
| | - Xinying Li
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
- School of Public Health, Shantou University, Shantou, Guangdong 515063, China
| | - Yanqi Wang
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
- School of Public Health, Shantou University, Shantou, Guangdong 515063, China
| | - Xiaojuan Chen
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
- School of Public Health, Shantou University, Shantou, Guangdong 515063, China
| | - Xiaojie Wang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Jun Luo
- Department of Cardiovascular Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Lifang Chen
- Department of Cardiovascular Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Jun Wu
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518000, China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518000, China
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518000, China
| | - Jianping Ma
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| | - Pei Qin
- Center for Clinical Epidemiology and Evidence-Based Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong 518000, China
| |
Collapse
|
3
|
Shukla A, Sharma C, Malik MZ, Singh AK, Aditya AK, Mago P, Shalimar, Ray AK. Deciphering the tripartite interaction of urbanized environment, gut microbiome and cardio-metabolic disease. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 377:124693. [PMID: 40022791 DOI: 10.1016/j.jenvman.2025.124693] [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: 12/02/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
The world is experiencing a sudden surge in urban population, especially in developing Asian and African countries. Consequently, the global burden of cardio-metabolic disease (CMD) is also rising owing to gut microbiome dysbiosis due to urbanization factors such as mode of birth, breastfeeding, diet, environmental pollutants, and soil exposure. Dysbiotic gut microbiome indicated by altered Firmicutes to Bacteroides ratio and loss of beneficial short-chain fatty acids-producing bacteria such as Prevotella, and Ruminococcus may disrupt host-intestinal homeostasis by altering host immune response, gut barrier integrity, and microbial metabolism through altered T-regulatory cells/T-helper cells balance, activation of pattern recognition receptors and toll-like receptors, decreased mucus production, elevated level of trimethylamine-oxide and primary bile acids. This leads to a pro-inflammatory gut characterized by increased pro-inflammatory cytokines such as tumour necrosis factor-α, interleukin-2, Interferon-ϒ and elevated levels of metabolites or metabolic endotoxemia due to leaky gut formation. These pathophysiological characteristics are associated with an increased risk of cardio-metabolic disease. This review aims to comprehensively elucidate the effect of urbanization on gut microbiome-driven cardio-metabolic disease. Additionally, it discusses targeting the gut microbiome and its associated pathways via strategies such as diet and lifestyle modulation, probiotics, prebiotics intake, etc., for the prevention and treatment of disease which can potentially be integrated into clinical and professional healthcare settings.
Collapse
Affiliation(s)
- Avaneesh Shukla
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Chanchal Sharma
- Department of Environmental Studies, University of Delhi, New Delhi, India
| | - Md Zubbair Malik
- Department of Translational Medicine, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Alok Kumar Singh
- Department of Zoology, Ramjas College, University of Delhi, New Delhi, India
| | - Abhishek Kumar Aditya
- Department of Medicine, K.D. Medical College, Hospital and Research Center, Mathura, India
| | - Payal Mago
- Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, New Delhi, India; Campus of Open Learning, University of Delhi, New Delhi, India
| | - Shalimar
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashwini Kumar Ray
- Department of Environmental Studies, University of Delhi, New Delhi, India.
| |
Collapse
|
4
|
Meulders B, Marei WFA, Loier L, Leroy JLMR. Lipotoxicity and Oocyte Quality in Mammals: Pathogenesis, Consequences, and Reversibility. Annu Rev Anim Biosci 2025; 13:233-254. [PMID: 39565833 DOI: 10.1146/annurev-animal-111523-102249] [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: 11/22/2024]
Abstract
Metabolic stress conditions are often characterized by upregulated lipolysis and subsequently increased serum free fatty acid (FFA) concentrations, leading to the uptake of FFAs by non-adipose tissues and impairment of their function. This phenomenon is known as lipotoxicity. The increased serum FFA concentrations are reflected in the ovarian follicular fluid, which can have harmful effects on oocyte development. Several studies using in vitro and in vivo mammalian models showed that altered oocyte metabolism, increased oxidative stress, and mitochondrial dysfunction are crucial mechanisms underlying this detrimental impact. Ultimately, this can impair offspring health through the persistence of defective mitochondria in the embryo, hampering epigenetic reprogramming and early development. In vitro and in vivo treatments to enhance oocyte mitochondrial function are increasingly being developed. This can help to improve pregnancy rates and safeguard offspring health in metabolically compromised individuals.
Collapse
Affiliation(s)
- Ben Meulders
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium; , , ,
| | - Waleed F A Marei
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium; , , ,
| | - Lien Loier
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium; , , ,
| | - Jo L M R Leroy
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium; , , ,
| |
Collapse
|
5
|
Kaylan KB, Nargis T, Figatner K, Wang JE, Pratuangtham S, Chakraborty A, Casimiro I, Nadler JL, Boxer MB, Maloney DJ, Anderson RM, Mirmira RG, Tersey SA. 12-Lipoxygenase inhibition improves glucose homeostasis and obesity-associated inflammation in human gene replacement mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632274. [PMID: 39868153 PMCID: PMC11761697 DOI: 10.1101/2025.01.10.632274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Obesity-associated inflammation is characterized by macrophage infiltration into peripheral tissues, contributing to the progression of prediabetes and type 2 diabetes (T2D). The enzyme 12-lipoxygenase (12-LOX) catalyzes the formation of pro-inflammatory eicosanoids and is known to promote the migration of macrophages, yet its role in obesity-associated inflammation remains incompletely understood. Furthermore, differences between mouse and human orthologs of 12-LOX have limited efforts to study existing pharmacologic inhibitors of 12-LOX. In this study, we utilized a human gene replacement mouse model in which the gene encoding mouse 12-LOX (Alox15) is replaced by the human ALOX12 gene. As a model of obesity and dysglycemia, we administered these mice a high-fat diet. We subsequently investigated the effects of VLX-1005, a potent and selective small molecule inhibitor of human 12-LOX. Oral administration of VLX-1005 resulted in improved glucose homeostasis, decreased β cell dedifferentiation, and reduced macrophage infiltration in islets and adipose tissue. Analysis of the stromal vascular fraction from adipose tissue showed a reduction in myeloid cells and cytokine expression with VLX-1005 treatment, indicating decreased adipose tissue inflammation. In a distinct mouse model in which Alox15 was selectively deleted in myeloid cells, we observed decreased β cell dedifferentiation and reduced macrophage infiltration in both islets and adipose tissue, suggesting that the effects of VLX-1005 may relate to the inhibition of 12-LOX in macrophages. These findings highlight 12-LOX as a key factor in obesity-associated inflammation and suggest that 12-LOX inhibition could serve as a therapeutic strategy to improve glucose homeostasis and peripheral inflammation in the setting of obesity and T2D.
Collapse
Affiliation(s)
- Kerim B. Kaylan
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Titli Nargis
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Kayla Figatner
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jiayi E. Wang
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarida Pratuangtham
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Advaita Chakraborty
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Isabel Casimiro
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Jerry L. Nadler
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | - Ryan M. Anderson
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Raghavendra G. Mirmira
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah A. Tersey
- Department of Medicine and Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
6
|
Sarma MK, Saucedo A, Sadananthan SA, Darwin CH, Felker ER, Raman S, Velan SS, Thomas MA. Lipid Deposition in Skeletal Muscle Tissues and Its Correlation with Intra-Abdominal Fat: A Pilot Investigation in Type 2 Diabetes Mellitus. Metabolites 2025; 15:25. [PMID: 39852368 PMCID: PMC11767081 DOI: 10.3390/metabo15010025] [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: 09/17/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: This study evaluated metabolites and lipid composition in the calf muscles of Type 2 diabetes mellitus (T2DM) patients and age-matched healthy controls using multi-dimensional MR spectroscopic imaging. We also explored the association between muscle metabolites, lipids, and intra-abdominal fat in T2DM. Methods: Participants included 12 T2DM patients (60.3 ± 8.6 years), 9 age-matched healthy controls (AMHC) (60.9 ± 7.8 years), and 10 young healthy controls (YHC) (28.3 ± 1.8 years). We acquired the 2D MR spectra of calf muscles using an enhanced accelerated 5D echo-planar correlated spectroscopic imaging (EP-COSI) technique and abdominal MRI with breath-hold 6-point Dixon sequence. Results: In YHC, choline levels were lower in the gastrocnemius (GAS) and soleus (SOL) muscles but higher in the tibialis anterior (TA) compared to AMHC. YHC also showed a higher unsaturation index (U.I.) of extramyocellular lipids (EMCL) in TA, intramyocellular lipids (IMCL) in GAS, carnosine in SOL, and taurine and creatine in TA. T2DM patients exhibited higher choline in TA and myo-inositol in SOL than AMHC, while triglyceride fat (TGFR2) levels in TA were lower. Correlation analyses indicated associations between IMCL U.I. and various metabolites in muscles with liver, pancreas, and abdominal fat estimates in T2DM. Conclusions: This study highlights distinct muscle metabolite and lipid composition patterns across YHC, AMHC, and T2DM subjects. Associations between IMCL U.I. and abdominal fat depots underscore the interplay between muscle metabolism and adiposity in T2DM. These findings provide new insights into metabolic changes in T2DM and emphasize the utility of advanced MR spectroscopic imaging in characterizing muscle-lipid interactions.
Collapse
Affiliation(s)
- Manoj Kumar Sarma
- Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.K.S.); (E.R.F.); (S.R.)
| | - Andres Saucedo
- Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.K.S.); (E.R.F.); (S.R.)
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential, Agency for Science, Technology, and Research (A*STAR), Singapore 117609, Singapore; (S.A.S.); (S.S.V.)
| | | | - Ely Richard Felker
- Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.K.S.); (E.R.F.); (S.R.)
| | - Steve Raman
- Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.K.S.); (E.R.F.); (S.R.)
| | - S. Sendhil Velan
- Institute for Human Development and Potential, Agency for Science, Technology, and Research (A*STAR), Singapore 117609, Singapore; (S.A.S.); (S.S.V.)
| | - Michael Albert Thomas
- Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.K.S.); (E.R.F.); (S.R.)
| |
Collapse
|
7
|
Samant V, Prabhu A. Exercise, exerkines and exercise mimetic drugs: Molecular mechanisms and therapeutics. Life Sci 2024; 359:123225. [PMID: 39522716 DOI: 10.1016/j.lfs.2024.123225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic diseases linked with sedentary lifestyles and poor dietary habits are increasingly common in modern society. Exercise is widely acknowledged to have a plethora of health benefits, including its role in primary prevention of various chronic conditions like type 2 diabetes mellitus, obesity, cardiovascular disease, and several musculoskeletal as well as degenerative disorders. Regular physical activity induces numerous physiological adaptations that contribute to these positive effects, primarily observed in skeletal muscle but also impacting other tissues. There is a growing interest among researchers in developing pharmaceutical interventions that mimic the beneficial effects of exercise for therapeutic applications. Exercise mimetic medications have the potential to be helpful aids in enhancing functional outcomes for patients with metabolic dysfunction, neuromuscular and musculoskeletal disorders. Some of the potential targets for exercise mimetics include pathways involved in metabolism, mitochondrial function, inflammation, and tissue regeneration. The present review aims to provide an exhaustive overview of the current understanding of exercise physiology, the role of exerkines and biomolecular pathways, and the potential applications of exercise mimetic drugs for the treatment of several diseases.
Collapse
Affiliation(s)
- Vedant Samant
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
8
|
Jin M, Shen Y, Monroig Ó, Zhao W, Bao Y, Zhu T, Tocher DR, Zhou Q. Sirt1 Mitigates Hepatic Lipotoxic Injury Induced by High-Fat-Diet in Fish Through Ire1α Deacetylation. J Nutr 2024; 154:3210-3224. [PMID: 39303797 DOI: 10.1016/j.tjnut.2024.09.013] [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: 05/05/2024] [Revised: 08/07/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Silent information regulator protein 1 (Sirt1) is crucial in regulating lipid metabolism, but its specific role and mechanism in fish hepatic lipotoxic injury remain undefined. OBJECTIVES This study aimed to elucidate the regulatory role of Sirt1 and the underlying mechanisms in dietary lipid-induced hepatic lipotoxic injury in a marine teleost black seabream. METHODS Black seabream were fed a control diet (12% lipid level), high-fat diet (HFD) [18% lipid level, oleic acid (OA)-rich], or HFD supplemented with 0.25%, 0.50%, or 1.00% resveratrol (RSV) for 8 wk. The cultured hepatocytes were stimulated by OA (200 μM), OA supplemented with RSV (20 μM), or transfection with sirt1-small interfering RNA (sisirt1). Biochemical indices, gene expression (qPCR), histology, transmission electron microscope, immunofluorescence, Western blot, flow cytometry, and immunoprecipitation assays were conducted to evaluate hepatic lipid deposition, lipid metabolism, endoplasmic reticulum stress, inflammation and apoptosis, and determine protein interactions between Sirt1 and Ire1α. RESULTS In vivo, RSV supplementation increased mRNA and protein expression levels of sirt1 (236.2% ± 16.1% and 53.1% ± 14.3%) and downregulated the mRNA and phosphorylated protein expression levels of ire1α/Ire1α (46.0% ± 7.6% and 38.6% ± 7.0%), jnk/Jnk (57.6% ± 7.3% and 122.1%), and nuclear factor κ B (nf-κb/Nf-κb) p65 (41.7% ± 7.1% and 24.6% ± 0.8%) compared with the HFD group. Similar patterns were found in the in vitro experiments; however, after knockdown of sirt1, although the cells were incubated with RSV, the expression levels of ire1α/ Ire1α, jnk/Jnk, and nf-κb/Nf-κb p65 showed no significant differences compared with the OA treatment. Moreover, we found that mutation of K61 to arginine to mimic Ire1α deacetylation confers protection against Ire1α-mediated OA-rich HFD-induced inflammation and apoptosis. CONCLUSIONS The findings revealed that Sirt1 protects against OA-rich HFD-induced hepatic lipotoxic injury via the deacetylation of Ire1α on K61, hence reducing Ire1α autophosphorylation level, and suppressing Jnk and Nf-κb p65 activation. This mechanism is elucidated for the first time in fish.
Collapse
Affiliation(s)
- Min Jin
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, China; Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo, China
| | - Yuedong Shen
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, China; Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo, China.
| | - Óscar Monroig
- Instituto de Acuicultura Torre de la Sal (IATS), CSIC, 12595 Ribera de Cabanes, Castellon, Spain
| | - Wenli Zhao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, China; Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo, China
| | - Yangguang Bao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, China; Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo, China
| | - Tingting Zhu
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, China; Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo, China
| | - Douglas R Tocher
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, China
| | - Qicun Zhou
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, China; Key Laboratory of Aquaculture Biotechnology Ministry of Education, Ningbo University, Ningbo, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo, China.
| |
Collapse
|
9
|
Fan MW, Tian JL, Chen T, Zhang C, Liu XR, Zhao ZJ, Zhang SH, Chen Y. Role of cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes pathway in diabetes and its complications. World J Diabetes 2024; 15:2041-2057. [PMID: 39493568 PMCID: PMC11525733 DOI: 10.4239/wjd.v15.i10.2041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Diabetes mellitus (DM) is one of the major causes of mortality worldwide, with inflammation being an important factor in its onset and development. This review summarizes the specific mechanisms of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway in mediating inflammatory responses. Furthermore, it comprehensively presents related research progress and the subsequent involvement of this pathway in the pathogenesis of early-stage DM, diabetic gastroenteropathy, diabetic cardiomyopathy, non-alcoholic fatty liver disease, and other complications. Additionally, the role of cGAS-STING in autonomic dysfunction and intestinal dysregulation, which can lead to digestive complications, has been discussed. Altogether, this study provides a comprehensive analysis of the research advances regarding the cGAS-STING pathway-targeted therapeutic agents and the prospects for their application in the precision treatment of DM.
Collapse
Affiliation(s)
- Ming-Wei Fan
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Jin-Lan Tian
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Tan Chen
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Can Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Xin-Ru Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Zi-Jian Zhao
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Shu-Hui Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| | - Yan Chen
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou 256600, Shandong Province, China
| |
Collapse
|
10
|
Djousse L, Zhou X, Lim J, Kim E, Sesso HD, Lee IM, Buring JE, McClelland RL, Gaziano JM, Steffen LM, Manson JE. Potato Consumption and Risk of Type 2 Diabetes Mellitus: A Harmonized Analysis of 7 Prospective Cohorts. J Nutr 2024; 154:3079-3087. [PMID: 39289134 DOI: 10.1016/j.tjnut.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/27/2024] [Accepted: 07/12/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Data on the relation of potato consumption with risk of type 2 diabetes (T2D) are limited and inconsistent. It is unclear whether the plant-based diet index (PDI), which is a novel and comprehensive tool to assess overall dietary pattern, modifies the association of potato intake with T2D. OBJECTIVES We examined the association of total, combined baked, boiled, and mashed potatoes and fried potatoes with risk of T2D and test the interaction between PDI score and potato consumption on T2D risk. METHODS We conducted a de novo, harmonized, individual-level data from 7 United States cohorts (N = 105,531). Cox regression was used to estimate hazard ratios (HRs) separately in each cohort adjusting for anthropometric, demographic, and lifestyle factors and cohort-specific results were pooled using an inverse-variance weighted method. RESULTS Mean age ranged from 25 to 72 y, 65% women, and mean consumption of total potatoes ranged from 1.9 to 4.3 times per week. In the primary analysis, total potato intake was not associated with T2D risk: multivariable adjusted HR of 1.01 (95% confidence interval [CI]: 0.95, 1.08) for consumption of 1-2 servings/wk; 1.01 (95% CI: 0.93, 1.10) for >2-3 servings/wk; 1.05 (95% CI: 0.99, 1.12) for >3 to <5 servings/wk; and 1.07 (95% CI: 0.99, 1.16) for 5+ servings/wk compared with no potato intake. In secondary analyses, consumption of combined baked, boiled, and mashed potatoes was not associated with T2D risk, whereas fried potato consumption was positively associated with T2D risk: HR were 1 (ref), 1.07 (95% CI: 1.02, 1.12), and 1.12 (95% CI: 1.03, 1.22) for intake frequency of 0/wk, >0 to 1/wk, and >1/wk, respectively (P-trend = 0.04). There was no significant interaction between PDI score and potato consumption on T2D risk. CONCLUSIONS Although consumption of total potato is not associated with T2D risk, a modest elevated risk of T2D is observed with fried potato consumption.
Collapse
Affiliation(s)
- Luc Djousse
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States.
| | - Xia Zhou
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Jaewon Lim
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Eunjung Kim
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Howard D Sesso
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - I-Min Lee
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Julie E Buring
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Robyn L McClelland
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - John M Gaziano
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Qi L, Groeger M, Sharma A, Goswami I, Chen E, Zhong F, Ram A, Healy K, Hsiao EC, Willenbring H, Stahl A. Adipocyte inflammation is the primary driver of hepatic insulin resistance in a human iPSC-based microphysiological system. Nat Commun 2024; 15:7991. [PMID: 39266553 PMCID: PMC11393072 DOI: 10.1038/s41467-024-52258-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
Interactions between adipose tissue, liver and immune system are at the center of metabolic dysfunction-associated steatotic liver disease and type 2 diabetes. To address the need for an accurate in vitro model, we establish an interconnected microphysiological system (MPS) containing white adipocytes, hepatocytes and proinflammatory macrophages derived from isogenic human induced pluripotent stem cells. Using this MPS, we find that increasing the adipocyte-to-hepatocyte ratio moderately affects hepatocyte function, whereas macrophage-induced adipocyte inflammation causes lipid accumulation in hepatocytes and MPS-wide insulin resistance, corresponding to initiation of metabolic dysfunction-associated steatotic liver disease. We also use our MPS to identify and characterize pharmacological intervention strategies for hepatic steatosis and systemic insulin resistance and find that the glucagon-like peptide-1 receptor agonist semaglutide improves hepatocyte function by acting specifically on adipocytes. These results establish our MPS modeling the adipose tissue-liver axis as an alternative to animal models for mechanistic studies or drug discovery in metabolic diseases.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Marko Groeger
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Aditi Sharma
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ishan Goswami
- Department of Bioengineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Erzhen Chen
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Fenmiao Zhong
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Apsara Ram
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kevin Healy
- Department of Bioengineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Materials Science and Engineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Edward C Hsiao
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Holger Willenbring
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- Liver Center, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
12
|
Liu J, Luo Y, Zhu YR, Wu ZT, Hou Y, Xu YJ, Li L, Ma CW. Correlation between type 2 diabetes mellitus remission and intrapancreatic fat deposition. World J Clin Cases 2024; 12:4536-4542. [PMID: 39070807 PMCID: PMC11235486 DOI: 10.12998/wjcc.v12.i21.4536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/09/2024] [Accepted: 06/13/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Intrapancreatic fat deposition (IPFD) exerts a significant negative impact on patients with type 2 diabetes mellitus (T2DM), accelerates disease deterioration, and may lead to impaired β-cell quality and function. AIM To investigate the correlation between T2DM remission and IPFD. METHODS We enrolled 80 abdominally obese patients with T2DM admitted to our institution from January 2019 to October 2023, including 40 patients with weight loss-induced T2DM remission (research group) and 40 patients with short-term intensive insulin therapy-induced T2DM remission (control group). We comparatively analyzed improvements in IPFD [differential computed tomography (CT) values of the spleen and pancreas and average CT value of the pancreas]; levels of fasting blood glucose (FBG), 2-h postprandial blood glucose (2hPBG), and insulin; and homeostasis model assessment of insulin resistance (HOMA-IR) scores. Correlation analysis was performed to explore the association between T2DM remission and IPFD. RESULTS After treatment, the differential CT values of the spleen and pancreas, FBG, 2hPBG, and HOMA-IR in the research group were significantly lower than those before treatment and in the control group, and the average CT value of the pancreas and insulin levels were significantly higher. Correlation analysis revealed that the greater the T2DM remission, the lower the amount of IPFD. CONCLUSION T2DM remission and IPFD are inversely correlated.
Collapse
Affiliation(s)
- Jiang Liu
- Division of Endocrinology, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Yue Luo
- Department of Radiological Medicine, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Yin-Rong Zhu
- Division of Endocrinology, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Zu-Tao Wu
- Division of Endocrinology, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Yan Hou
- Division of Endocrinology, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Yu-Jie Xu
- Division of Endocrinology, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Li Li
- Division of Endocrinology, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| | - Cun-Wen Ma
- Department of Radiological Medicine, Wenshan Hospital Affiliated to Kunming University of Science and Technology, Wenshan 663000, Yunnan Province, China
| |
Collapse
|
13
|
Wang H, Wang J, Cui H, Fan C, Xue Y, Liu H, Li H, Li J, Li H, Sun Y, Wang W, Song J, Jiang C, Xu M. Inhibition of fatty acid uptake by TGR5 prevents diabetic cardiomyopathy. Nat Metab 2024; 6:1161-1177. [PMID: 38698281 PMCID: PMC11199146 DOI: 10.1038/s42255-024-01036-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Diabetic cardiomyopathy is characterized by myocardial lipid accumulation and cardiac dysfunction. Bile acid metabolism is known to play a crucial role in cardiovascular and metabolic diseases. Takeda G-protein-coupled receptor 5 (TGR5), a major bile acid receptor, has been implicated in metabolic regulation and myocardial protection. However, the precise involvement of the bile acid-TGR5 pathway in maintaining cardiometabolic homeostasis remains unclear. Here we show decreased plasma bile acid levels in both male and female participants with diabetic myocardial injury. Additionally, we observe increased myocardial lipid accumulation and cardiac dysfunction in cardiomyocyte-specific TGR5-deleted mice (both male and female) subjected to a high-fat diet and streptozotocin treatment or bred on the diabetic db/db genetic background. Further investigation reveals that TGR5 deletion enhances cardiac fatty acid uptake, resulting in lipid accumulation. Mechanistically, TGR5 deletion promotes localization of CD36 on the plasma membrane through the upregulation of CD36 palmitoylation mediated by the palmitoyl acyltransferase DHHC4. Our findings indicate that the TGR5-DHHC4 pathway regulates cardiac fatty acid uptake, which highlights the therapeutic potential of targeting TGR5 in the management of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hu Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China
| | - Jiaxing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China
| | - Hao Cui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Chenyu Fan
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China
| | - Yuzhou Xue
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China
| | - Huiying Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hui Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Houhua Li
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, China.
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
14
|
Kim EJ, Jeon HB, Kang MJ, Lee J. Dynamic Imaging of Lipid Droplets in Cells and Tissues by Using Dioxaborine Barbiturate-Based Fluorogenic Probes. Anal Chem 2024; 96:8356-8364. [PMID: 38753674 DOI: 10.1021/acs.analchem.3c05368] [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: 05/18/2024]
Abstract
Lipids are essential for various cellular functions, including energy storage, membrane flexibility, and signaling molecule production. Maintaining proper lipid levels is important to prevent health problems such as cancer, neurodegenerative disorders, cardiovascular diseases, obesity, and diabetes. Monitoring cellular lipid droplets (LDs) in real-time with high resolution can provide insights into LD-related pathways and diseases owing to the dynamic nature of LDs. Fluorescence-based imaging is widely used for tracking LDs in live cells and animal models. However, the current fluorophores have limitations such as poor photostability and high background staining. Herein, we developed a novel fluorogenic probe based on a push-pull interaction combined with aggregation-induced emission enhancement (AIEE) for dynamic imaging of LDs. Probe 1 exhibits favorable membrane permeability and spectroscopic characteristics, allowing specific imaging of cellular LDs and time-lapse imaging of LD accumulation. This probe can also be used to examine LDs in fruit fly tissues in various metabolic states, serving as a highly versatile and specific tool for dynamic LD imaging in cellular and tissue environments.
Collapse
Affiliation(s)
- Eun-Ji Kim
- Department of Next-Generation Applied Science and School of Biopharmaceutical and Medical Sciences, Sungshin University, Seoul 01133, Republic of Korea
| | - Hye-Bin Jeon
- Department of Next-Generation Applied Science and School of Biopharmaceutical and Medical Sciences, Sungshin University, Seoul 01133, Republic of Korea
| | - Min-Ji Kang
- Department of Pharmacology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Jiyoun Lee
- Department of Next-Generation Applied Science and School of Biopharmaceutical and Medical Sciences, Sungshin University, Seoul 01133, Republic of Korea
| |
Collapse
|
15
|
Abdoon ASS, Hegazy AM, Abdel-Azeem AS, Al-Atrash AM, Mohammed DM. The protective effects of some herbs on mitigating HFD-induced obesity via enhancing biochemical indicators and fertility in female rats. Heliyon 2024; 10:e30249. [PMID: 38726161 PMCID: PMC11078881 DOI: 10.1016/j.heliyon.2024.e30249] [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: 01/04/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
The potential of plant-based diets and drugs to prevent and control obesity has been attributed to the presence of several biologically active phytochemicals. The study aimed to assess herb consumption's impact on alleviating the risks and hazards associated with obesity induced by a high-fat diet (HFD) and the promotion of fertility. Eighty rats were allocated into four distinct groups. Group 1 (G1) was provided with a basal diet and acted as the control group. Group 2 (G2) was provided with an HFD. Group 3 (G3) was provided with HFD supplemented with chia seeds and Hibiscus sabdariffa L. The fourth group of subjects was provided with HFD supplemented with Foeniculum vulgare (fennel) and Coriandrum sativum L. (coriander). The feeding session was sustained for 10 weeks, and the biochemical parameters were evaluated. The administration of Foeniculum vulgare (fennel) and Coriandrum sativum L. (coriander) (G4) resulted in a more significant reduction in all biochemical parameters compared to G3, which received a diet consisting of chia seeds and Hibiscus sabdariffa L. Additionally, the average number of embryonic lobes and the average number of offspring after birth were found to be considerably more significant in the normal control group (G1) and group (G4) compared to the HFD group (G2) and group (G3) (P < 0.01). Group 4 (G4) was administered a diet enriched with Foeniculum vulgare (fennel) and Coriandrum sativum L. (coriander), which demonstrated superior outcomes in many biochemical indicators and the promotion of fertility in obese female rats.
Collapse
Affiliation(s)
- Ahmed Sabry S. Abdoon
- Animal Reproduction, Veterinary Research division, National Research Centre, Dokki, Giza, Egypt
| | - Amany M. Hegazy
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, Egypt
| | - Amal S. Abdel-Azeem
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, Egypt
| | - Ahmed M. Al-Atrash
- Medical Administration, Nuclear Materials Authority, Kattmya, Cairo, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
16
|
Al-Rashed F, Arefanian H, Madhoun AA, Bahman F, Sindhu S, AlSaeed H, Jacob T, Thomas R, Al-Roub A, Alzaid F, Malik MDZ, Nizam R, Thanaraj TA, Al-Mulla F, Hannun YA, Ahmad R. Neutral Sphingomyelinase 2 Inhibition Limits Hepatic Steatosis and Inflammation. Cells 2024; 13:463. [PMID: 38474427 PMCID: PMC10931069 DOI: 10.3390/cells13050463] [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: 01/24/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is manifested by hepatic steatosis, insulin resistance, hepatocyte death, and systemic inflammation. Obesity induces steatosis and chronic inflammation in the liver. However, the precise mechanism underlying hepatic steatosis in the setting of obesity remains unclear. Here, we report studies that address this question. After 14 weeks on a high-fat diet (HFD) with high sucrose, C57BL/6 mice revealed a phenotype of liver steatosis. Transcriptional profiling analysis of the liver tissues was performed using RNA sequencing (RNA-seq). Our RNA-seq data revealed 692 differentially expressed genes involved in processes of lipid metabolism, oxidative stress, immune responses, and cell proliferation. Notably, the gene encoding neutral sphingomyelinase, SMPD3, was predominantly upregulated in the liver tissues of the mice displaying a phenotype of steatosis. Moreover, nSMase2 activity was elevated in these tissues of the liver. Pharmacological and genetic inhibition of nSMase2 prevented intracellular lipid accumulation and TNFα-induced inflammation in in-vitro HepG2-steatosis cellular model. Furthermore, nSMase2 inhibition ameliorates oxidative damage by rescuing PPARα and preventing cell death associated with high glucose/oleic acid-induced fat accumulation in HepG2 cells. Collectively, our findings highlight the prominent role of nSMase2 in hepatic steatosis, which could serve as a potential therapeutic target for NAFLD and other hepatic steatosis-linked disorders.
Collapse
Affiliation(s)
- Fatema Al-Rashed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Hossein Arefanian
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Ashraf Al Madhoun
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (S.S.)
| | - Fatemah Bahman
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Sardar Sindhu
- Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (S.S.)
| | - Halemah AlSaeed
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Texy Jacob
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Reeby Thomas
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Areej Al-Roub
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| | - Fawaz Alzaid
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France;
| | - MD Zubbair Malik
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Thangavel Alphonse Thanaraj
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (M.Z.M.); (R.N.); (T.A.T.); (F.A.-M.)
| | - Yusuf A. Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Rasheed Ahmad
- Immunology & Microbiology Department, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.B.); (H.A.); (T.J.); (R.T.); (A.A.-R.)
| |
Collapse
|
17
|
Lu T, Kan J, He X, Zou J, Sheng D, Xue Y, Wang Y, Xu L. Gastric Submucosal Fat Accumulation Is Associated with Insulin Resistance in Patients with Obesity. Obes Surg 2024; 34:534-541. [PMID: 38191965 PMCID: PMC10811089 DOI: 10.1007/s11695-023-07014-2] [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: 07/25/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE Ectopic fat accumulation plays a significant role in obesity-related metabolic dysfunction, and few studies have reported an association between ectopic gastric fat and metabolic risk factors. We aim to fulfill this need by assessing the degree of gastric submucosal fat accumulation in pathologic sections of 190 sleeve gastrectomy specimens. METHODS Study patients were divided into two groups (D1 and D2) based on whether fat accumulation exceeded 1/3 of the submucosa of the stomach. Demographic and metabolic risk factors were compared between the two groups. Metabolic risk variables that might be associated with the degree of fat accumulation were screened in the original cohort. After balancing for possible confounders, the robustness of the correlations was assessed using binary and conditional logistic regression analyses. RESULTS All study patients had fat accumulation in the submucosa of the stomach. C-reactive protein (CRP), body mass index (BMI), visceral fat area (VFA), and insulin resistance (IR) were higher in the D2 group than in the D1 group in the original cohort (P < 0.05). Logistic regression analysis showed that BMI and IR may be associated with increased fat accumulation. After balancing variables other than obesity indicators and IR using propensity score matching, BMI and IR remained significantly different between the two groups (P < 0.05). Further analysis of the matched cohort using two logistic regression analyses showed that IR was an independent risk factor for increased fat accumulation. CONCLUSION This study indicated that gastric submucosal fat accumulation was prevalent in patients with obesity and was associated with IR.
Collapse
Affiliation(s)
- Tao Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Jianxun Kan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Xue He
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Jialai Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Dandan Sheng
- Department of Nuclear Medicine, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Yating Xue
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Yan Wang
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China
| | - Lijian Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiang Jia Yuan Road, Nanjing, 210011, Jiangsu Province, China.
| |
Collapse
|
18
|
An Q, Zhang QH, Wang Y, Zhang HY, Liu YH, Zhang ZT, Zhang ML, Lin LJ, He H, Yang YF, Sun P, Zhou ZY, Song QW, Liu AL. Association between type 2 diabetes mellitus and body composition based on MRI fat fraction mapping. Front Public Health 2024; 12:1332346. [PMID: 38322122 PMCID: PMC10846073 DOI: 10.3389/fpubh.2024.1332346] [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: 11/02/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Purpose To explore the association between type 2 diabetes mellitus (T2DM) and body composition based on magnetic resonance fat fraction (FF) mapping. Methods A total of 341 subjects, who underwent abdominal MRI examination with FF mapping were enrolled in this study, including 68 T2DM patients and 273 non-T2DM patients. The FFs and areas of visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT) and abdominal muscle (AM) were measured at the level of the L1-L2 vertebral. The FF of bone marrow adipose tissue (BMAT) was determined by the averaged FF values measured at the level of T12 and L1 vertebral, respectively. The whole hepatic fat fraction (HFF) and pancreatic fat fraction (PFF) were measured based on 3D semi-automatic segmentation on the FF mapping. All data were analyzed by GraphPad Prism and MedCalc. Results VAT area, VAT FF, HFF, PFF of T2DM group were higher than those of non-T2DM group after adjusting for age and sex (P < 0.05). However, there was no differences in SAT area, SAT FF, BMAT FF, AM area and AM FF between the two groups (P > 0.05). VAT area and PFF were independent risk factors of T2DM (all P < 0.05). The area under the curve (AUC) of the receiver operating characteristic (ROC) for VAT area and PFF in differentiating between T2DM and non-T2DM were 0.685 and 0.787, respectively, and the AUC of PFF was higher than VAT area (P < 0.05). Additionally, in seemingly healthy individuals, the SAT area, VAT area, and AM area were found to be significantly associated with being overweight and/or obese (BMI ≥ 25) (all P < 0.05). Conclusions In this study, it was found that there were significant associations between T2DM and VAT area, VAT FF, HFF and PFF. In addition, VAT area and PFF were the independent risk factors of T2DM. Especially, PFF showed a high diagnostic performance in discrimination between T2DM and non-T2DM. These findings may highlight the crucial role of PFF in the pathophysiology of T2DM, and it might be served as a potential imaging biomarker of the prevention and treatment of T2DM. Additionally, in individuals without diabetes, focusing on SAT area, VAT area and AM area may help identify potential health risks and provide a basis for targeted weight management and prevention measures.
Collapse
Affiliation(s)
- Qi An
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qin-He Zhang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Han-Yue Zhang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu-Hui Liu
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | - Zi-Ting Zhang
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | - Mei-Ling Zhang
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | | | - Hui He
- Department of Thyroid, Metabolic Diseases and Hernia Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yi-Fan Yang
- Department of Thyroid, Metabolic Diseases and Hernia Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Peng Sun
- Philips Healthcare, Beijing, China
| | | | - Qing-Wei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ai-Lian Liu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
Zhou Q, Yan H, Jin A, Meng X, Lin J, Li H, Wang Y, Pan Y. Adipose tissue specific insulin resistance and prognosis of nondiabetic patients with ischemic stroke. Diabetol Metab Syndr 2023; 15:246. [PMID: 38041145 PMCID: PMC10693091 DOI: 10.1186/s13098-023-01235-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/25/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Insulin resistance is linked to atherosclerotic cardiovascular diseases and stroke, whereas less is known about adipose tissue specific insulin resistance and outcomes after ischemic stroke. This study aimed to estimate the association between adipose tissue specific insulin resistance and prognosis of nondiabetic patients with ischemic stroke. METHODS Patients with ischemic stroke without a history of diabetes mellitus in the Third China National Stroke Registry were included. Adipose tissue specific insulin resistance index (Adipo-IR) was calculated by fasting serum insulin and free fatty acids and categorized into 5 groups according to the quintiles. Outcomes included stroke recurrence (ischemic or hemorrhagic), combined vascular events, all-cause death, and poor outcome (modified Rankin Scale, 3-6) at 12 months after stroke onset. We assessed the association between Adipo-IR and risk of prognosis by multivariable Cox/logistic regression models adjusted for potential covariates. RESULTS Among 2,222 patients, 69.0% were men with a mean age of 62.5 years. At 12 months, 185 (8.3%) patients had recurrent stroke, 193 (8.7%) had combined vascular events, 58 (2.6%) died, and 250 (11.5%) had a poor outcome. Compared with patients with the lowest quintile, patients with the second, third, fourth, fifth quintiles of the Adipo-IR were associated with an increased risk of stroke recurrence (hazard ratio [HR], 1.77; 95% CI, 1.04-3.03; P = 0.04; HR, 2.19; 95% CI, 1.30-3.68; P = 0.003; HR, 1.84; 95% CI, 1.06-3.21; P = 0.03; HR, 2.11; 95% CI, 1.20-3.71; P = 0.01, respectively) and marginally associated with an increased risk of combined vascular events ( HR, 1.60; 95%CI, 0.97-2.64; P = 0.07; HR, 1.91; 95% CI, 1.17-3.13; P = 0.01; HR, 1.62; 95% CI, 0.96-2.75; P = 0.07; HR, 1.80; 95% CI, 1.05-3.09; P = 0.03, respectively) at 12 months after adjustment for potential covariates. Adipo-IR was not associated with mortality and poor outcome at 12 months. CONCLUSIONS These findings suggest that adipose tissue specific insulin resistance is independently associated with recurrent stroke and combined vascular events after acute ischemic stroke in nondiabetic patients.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hongyi Yan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Aoming Jin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jinxi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
20
|
Kim J, Kim NH, Youn I, Seo EK, Kim CY. Effects of Allium macrostemon Bunge Extract on Adipose Tissue Inflammation and Hepatic Endoplasmic Reticulum Stress in High-Fat Diet-Fed and Bisphenol A-Treated C57BL/6N Mice. Foods 2023; 12:3777. [PMID: 37893670 PMCID: PMC10606828 DOI: 10.3390/foods12203777] [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: 08/21/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The simultaneous exposure to a high-fat (HF) diet and to bisphenol A (BPA) from delivered foods and food-delivery containers is on the rise in humans, according to the increased frequency of food delivery during the COVID-19 pandemic. This co-exposure could cause harmful tissue toxicity in the human body. Here, the preventive effect of Allium macrostemon Bunge (AM) extract against dysfunction in adipose tissue and the liver under co-exposure to BPA and an HF diet was examined using mice. C57BL/6N mice were divided into four groups (n = 6 or 7/group) according to diet and treatment: control diet with vehicle (CON), HF diet with vehicle (HF), HF diet with an oral injection of BPA (HF + BP), and HF diet with an oral injection of BPA and AM extract (HF + BP + AM). HF feeding increased body weight gain compared to CON feeding, while BP + HF and BP + HF + AM feeding suppressed body weight gain compared with HF feeding. The BP + HF group had lower body weight than the HF group, but the two groups had similar epididymal fat mass. The HF + BP + AM group showed lower pro-inflammatory gene expression levels in adipose tissue and epididymal fat mass compared to the HF + BP group. Altered endoplasmic reticulum (ER) stress response in the liver was partly observed in the HF + BP group, as shown by increased total phosphorylated Jun N-terminal kinase protein levels compared to those in the HF group. In addition, ecdysterone 25-O-β-D-glucopyranoside and 6-gingerol were identified in AM extract by mass spectrometry and molecular networking analysis. In summary, the AM extract diminished adipose tissue inflammation and hepatic ER stress in an HF diet and BPA co-exposure condition. To utilize AM as a potential food component to alleviate the harmful effect of an HF diet and BPA exposure, further research investigating the specific impact of AM extract supplementation using additional experimental groups or various treatment doses is warranted.
Collapse
Affiliation(s)
- Juhae Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (J.K.); (N.-H.K.)
| | - Na-Hyung Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (J.K.); (N.-H.K.)
| | - Isoo Youn
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (I.Y.); (E.K.S.)
| | - Eun Kyoung Seo
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (I.Y.); (E.K.S.)
| | - Choon Young Kim
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (J.K.); (N.-H.K.)
- Department of Food and Nutrition, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
21
|
Xu S, Xi J, Wu T, Wang Z. The Role of Adipocyte Endoplasmic Reticulum Stress in Obese Adipose Tissue Dysfunction: A Review. Int J Gen Med 2023; 16:4405-4418. [PMID: 37789878 PMCID: PMC10543758 DOI: 10.2147/ijgm.s428482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
Adipose tissue dysfunction plays an important role in metabolic diseases associated with chronic inflammation, insulin resistance and lipid ectopic deposition in obese patients. In recent years, it has been found that under the stimulation of adipocyte endoplasmic reticulum stress (ERS), the over-activated ER unfolded protein response (UPR) exacerbates the inflammatory response of adipose tissue by interfering with the normal metabolism of adipose tissue, promotes the secretion of adipokines, and affects the browning and thermogenic pathways of adipose tissue, ultimately leading to the manifestation of metabolic syndrome such as ectopic lipid deposition and disorders of glucolipid metabolism in obese patients. This paper mainly summarizes the relationship between adipocyte ERS and obese adipose tissue dysfunction and provides an overview of the mechanisms by which ERS induces metabolic disorders such as catabolism, thermogenesis and inflammation in obese adipose tissue through the regulation of molecules and pathways such as NF-κB, ADPN, STAMP2, LPIN1, TRIP-Br2, NF-Y and SIRT2 and briefly describes the current mechanisms targeting adipocyte endoplasmic reticulum stress to improve obesity and provide ideas for intervention and treatment of obese adipose tissue dysfunction.
Collapse
Affiliation(s)
- Shengjie Xu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Jiaqiu Xi
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Tao Wu
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Zhonglin Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, People’s Republic of China
| |
Collapse
|
22
|
Xiao Y, Pietzner A, Rohwer N, Jung A, Rothe M, Weylandt KH, Elbelt U. Bioactive oxylipins in type 2 diabetes mellitus patients with and without hypertriglyceridemia. Front Endocrinol (Lausanne) 2023; 14:1195247. [PMID: 37664847 PMCID: PMC10472135 DOI: 10.3389/fendo.2023.1195247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/20/2023] [Indexed: 09/05/2023] Open
Abstract
Objective Dyslipidemia, in particular elevated triglycerides (TGs) contribute to increased cardiovascular risk in type 2 diabetes mellitus (T2DM). In this pilot study we aimed to assess how increased TGs affect hepatic fat as well as polyunsaturated fatty acid (PUFA) metabolism and oxylipin formation in T2DM patients. Methods 40 patients with T2DM were characterized analyzing routine lipid blood parameters, as well as medical history and clinical characteristics. Patients were divided into a hypertriglyceridemia (HTG) group (TG ≥ 1.7mmol/l) and a normal TG group with TGs within the reference range (TG < 1.7mmol/l). Profiles of PUFAs and their oxylipins in plasma were measured by gas chromatography and liquid chromatography/tandem mass spectrometry. Transient elastography (TE) was used to assess hepatic fat content measured as controlled attenuation parameter (CAP) (in dB/m) and the degree of liver fibrosis measured as stiffness (in kPa). Results Mean value of hepatic fat content measured as CAP as well as body mass index (BMI) were significantly higher in patients with high TGs as compared to those with normal TGs, and correlation analysis showed higher concentrations of TGs with increasing CAP and BMI scores in patients with T2DM. There were profound differences in plasma oxylipin levels between these two groups. Cytochrome P450 (CYP) and lipoxygenase (LOX) metabolites were generally more abundant in the HTG group, especially those derived from arachidonic acid (AA), eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), γ-linolenic acid (γ-LA), and α-linolenic acid (α-LA), and a strong correlation between TG levels and plasma metabolites from different pathways was observed. Conclusions In adult patients with T2DM, elevated TGs were associated with increased liver fat and BMI. Furthermore, these patients also had significantly higher plasma levels of CYP- and LOX- oxylipins, which could be a novel indicator of increased inflammatory pathway activity, as well as a novel target to dampen this activity.
Collapse
Affiliation(s)
- Yanan Xiao
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anne Pietzner
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
| | - Nadine Rohwer
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Adelheid Jung
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
| | | | - Karsten H. Weylandt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
| | - Ulf Elbelt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
23
|
Qi L, Matsuo K, Pereira A, Lee YT, Zhong F, He Y, Zushin PJH, Gröger M, Sharma A, Willenbring H, Hsiao EC, Stahl A. Human iPSC-Derived Proinflammatory Macrophages cause Insulin Resistance in an Isogenic White Adipose Tissue Microphysiological System. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2203725. [PMID: 37104853 PMCID: PMC10502939 DOI: 10.1002/smll.202203725] [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] [Received: 06/15/2022] [Revised: 02/01/2023] [Indexed: 06/08/2023]
Abstract
Chronic white adipose tissue (WAT) inflammation has been recognized as a critical early event in the pathogenesis of obesity-related disorders. This process is characterized by the increased residency of proinflammatory M1 macrophages in WAT. However, the lack of an isogenic human macrophage-adipocyte model has limited biological studies and drug discovery efforts, highlighting the need for human stem cell-based approaches. Here, human induced pluripotent stem cell (iPSC) derived macrophages (iMACs) and adipocytes (iADIPOs) are cocultured in a microphysiological system (MPS). iMACs migrate toward and infiltrate into the 3D iADIPOs cluster to form crown-like structures (CLSs)-like morphology around damaged iADIPOs, recreating classic histological features of WAT inflammation seen in obesity. Significantly more CLS-like morphologies formed in aged and palmitic acid-treated iMAC-iADIPO-MPS, showing the ability to mimic inflammatory severity. Importantly, M1 (proinflammatory) but not M2 (tissue repair) iMACs induced insulin resistance and dysregulated lipolysis in iADIPOs. Both RNAseq and cytokines analyses revealed a reciprocal proinflammatory loop in the interactions of M1 iMACs and iADIPOs. This iMAC-iADIPO-MPS thus successfully recreates pathological conditions of chronically inflamed human WAT, opening a door to study the dynamic inflammatory progression and identify clinically relevant therapies.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, California, 94720, USA
| | - Koji Matsuo
- Division of Endocrinology and Metabolism, Institute for Human Genetics, the Eli and Edythe Broad Institute for Regeneration Medicine, and the Program in Craniofacial Biology, Department of Medicine, University of California, San Francisco
| | - Ashley Pereira
- Division of Endocrinology and Metabolism, Institute for Human Genetics, the Eli and Edythe Broad Institute for Regeneration Medicine, and the Program in Craniofacial Biology, Department of Medicine, University of California, San Francisco
| | - Yue Tung Lee
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, California, 94720, USA
| | - Fenmiao Zhong
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, California, 94720, USA
| | - Yuchen He
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, California, 94720, USA
| | - Peter-James H. Zushin
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, California, 94720, USA
| | - Marko Gröger
- Division of Transplant Surgery, Department of Surgery; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; Liver Center, University of California, San Francisco
| | - Aditi Sharma
- Division of Endocrinology and Metabolism, Institute for Human Genetics, the Eli and Edythe Broad Institute for Regeneration Medicine, and the Program in Craniofacial Biology, Department of Medicine, University of California, San Francisco
| | - Holger Willenbring
- Division of Transplant Surgery, Department of Surgery; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; Liver Center, University of California, San Francisco
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Institute for Human Genetics, the Eli and Edythe Broad Institute for Regeneration Medicine, and the Program in Craniofacial Biology, Department of Medicine, University of California, San Francisco
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
24
|
Castera L, Cusi K. Diabetes and cirrhosis: Current concepts on diagnosis and management. Hepatology 2023; 77:2128-2146. [PMID: 36631005 DOI: 10.1097/hep.0000000000000263] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/03/2022] [Indexed: 01/13/2023]
Abstract
Type 2 diabetes mellitus is often associated with cirrhosis as comorbidities, acute illness, medications, and other conditions profoundly alter glucose metabolism. Both conditions are closely related in NAFLD, the leading cause of chronic liver disease, and given its rising burden worldwide, management of type 2 diabetes mellitus in cirrhosis will be an increasingly common dilemma. Having diabetes increases cirrhosis-related complications, including HCC as well as overall mortality. In the absence of effective treatments for cirrhosis, patients with type 2 diabetes mellitus should be systematically screened as early as possible for NAFLD-related fibrosis/cirrhosis using noninvasive tools, starting with a FIB-4 index followed by transient elastography, if available. In people with cirrhosis, an early diagnosis of diabetes is critical for an optimal management strategy (ie, nutritional goals, and glycemic targets). Diagnosis of diabetes may be missed if based on A1C in patients with cirrhosis and impaired liver function (Child-Pugh B-C) as anemia may turn the test unreliable. Clinicians must also become aware of their high risk of hypoglycemia, especially in decompensated cirrhosis where insulin is the only therapy. Care should be within multidisciplinary teams (nutritionists, obesity management teams, endocrinologists, hepatologists, and others) and take advantage of novel glucose-monitoring devices. Clinicians should become familiar with the safety and efficacy of diabetes medications for patients with advanced fibrosis and compensated cirrhosis. Management is conditioned by whether the patient has either compensated or decompensated cirrhosis. This review gives an update on the complex relationship between cirrhosis and type 2 diabetes mellitus, with a focus on its diagnosis and treatment, and highlights knowledge gaps and future directions.
Collapse
Affiliation(s)
- Laurent Castera
- Departement of Hepatology, Hospital Beaujon, Assistance Publique-Hôpitaux de Paris, INSERM UMR 1149, Université Paris Cité, Clichy, France
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, The University of Florida, Gainesville, Florida, USA
| |
Collapse
|
25
|
Sonkar SK, Gupta A, Sonkar GK, Usman K, Bhosale V, Kumar S, Sharma S. Zinc Alpha 2 Glycoprotein as an Early Biomarker of Diabetic Nephropathy in Type 2 Diabetes Mellitus Patients. Cureus 2023; 15:e36011. [PMID: 37051007 PMCID: PMC10085351 DOI: 10.7759/cureus.36011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2023] [Indexed: 03/13/2023] Open
Abstract
Background and objectives Microalbuminuria is an early sign of diabetic nephropathy (DN). However, pathological abnormalities occur before the onset of microalbuminuria. Renal impairment progresses in about 50% of cases in type 2 diabetes mellitus (T2DM) without significant albuminuria. Diabetes mellitus (DM) is linked with obesity, metabolic syndrome, and lifestyle changes, where adipokines play an important role. Zinc alpha 2 glycoprotein (ZAGP) is an adipokine, and in this study, it was assessed as a potential biomarker for early DN as well as its progression. Materials and methods This study was a cross-sectional case-control study conducted at a tertiary hospital in northern India. T2DM patients aged 18-65 years old were included in the study and were divided into four groups based on their albuminuria level. This study included 160 participants, with 40 participants in each group. Group I included healthy volunteers, while Groups II, III, and IV were normoalbuminuric, microalbuminuric, and macroalbuminuric diabetic patients, respectively. The groups were evaluated for demographic variables, biochemical parameters, urine albumin-creatinine ratio (UACR), and serum ZAGP. Data between the groups were compared statistically. Results This study included 160 participants, with 40 participants in each group. There was a significant difference between the groups based on the serum ZAGP (p<0.001). Serum ZAGP was significantly negatively correlated with serum creatinine, glycosylated hemoglobin (HbA1c), serum cholesterol, serum triglyceride, low-density lipoprotein (LDL) cholesterol, and UACR. ZAGP was positively correlated with the estimated glomerular filtration rate (eGFR). Conclusion The present study showed that ZAGP was an early biomarker of diabetic nephropathy, and its value decreased as DN progressed. It also suggested that ZAGP, an adipokine, has an anti-inflammatory mechanism of action and its depletion worsens the disease.
Collapse
|
26
|
Handy RM, DesOrmeaux GJ, Barbeau PA, Frangos SM, Holloway GP. Independent, but not co-supplementation, with nitrate and resveratrol improves glucose tolerance and reduces markers of cellular stress in high-fat-fed male mice. Am J Physiol Regul Integr Comp Physiol 2023; 324:R317-R328. [PMID: 36622081 DOI: 10.1152/ajpregu.00196.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Independent supplementation with nitrate (NIT) and resveratrol (RSV) enriches various aspects of mitochondrial biology in key metabolic tissues. Although RSV is known to activate Sirt1 and initiate mitochondrial biogenesis, the metabolic benefits elicited by dietary nitrate appear to be dependent on 5'-adenosine monophosphate-activated protein kinase (AMPK)-mediated signaling events, a process also linked to the activation of Sirt1. Although the benefits of individual supplementation with these compounds have been characterized, it is unknown if co-supplementation may produce superior metabolic adaptations. Thus, we aimed to determine if treatment with combined +NIT and +RSV (+RN) could additively alter metabolic adaptations in the presence of a high-fat diet (HFD). Both +RSV and +NIT improved glucose tolerance compared with HFD (P < 0.05); however, this response was attenuated following combined +RN supplementation. Within skeletal muscle, all supplements increased mitochondrial ADP sensitivity compared with HFD (P < 0.05), without altering mitochondrial content. Although +RSV and +NIT decreased hepatic lipid deposition compared with HFD (P < 0.05), this effect was abolished with +RN, which aligned with significant reductions in Sirt1 protein content (P < 0.05) after combined treatment, in the absence of changes to mitochondrial content or function. Within epididymal white adipose tissue (eWAT), all supplements reduced crown-like structure accumulation compared with HFD (P < 0.0001) and mitochondrial reactive oxygen species (ROS) emission (P < 0.05), alongside reduced adipocyte cross-sectional area (CSA) (P < 0.05), with the greatest effect observed after +RN treatment (P = 0.0001). Although the present data suggest additive changes in adipose tissue metabolism after +RN treatment, concomitant impairments in hepatic lipid homeostasis appear to prevent improvements in whole body glucose homeostasis observed with independent treatment, which may be Sirt1 dependent.
Collapse
Affiliation(s)
- Rachel M Handy
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Geneviève J DesOrmeaux
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Pierre-Andre Barbeau
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Sara M Frangos
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
27
|
Li G, Li X, Yang L, Wang S, Dai Y, Fekry B, Veillon L, Tan L, Berdeaux R, Eckel-Mahan K, Lorenzi PL, Zhao Z, Lehner R, Sun K. Adipose tissue-specific ablation of Ces1d causes metabolic dysregulation in mice. Life Sci Alliance 2022; 5:e202101209. [PMID: 35459739 PMCID: PMC9034061 DOI: 10.26508/lsa.202101209] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 01/25/2023] Open
Abstract
Carboxylesterase 1d (Ces1d) is a crucial enzyme with a wide range of activities in multiple tissues. It has been reported to localize predominantly in ER. Here, we found that Ces1d levels are significantly increased in obese patients with type 2 diabetes. Intriguingly, a high level of Ces1d translocates onto lipid droplets where it digests the lipids to produce a unique set of fatty acids. We further revealed that adipose tissue-specific Ces1d knock-out (FKO) mice gained more body weight with increased fat mass during a high fat-diet challenge. The FKO mice exhibited impaired glucose and lipid metabolism and developed exacerbated liver steatosis. Mechanistically, deficiency of Ces1d induced abnormally large lipid droplet deposition in the adipocytes, causing ectopic accumulation of triglycerides in other peripheral tissues. Furthermore, loss of Ces1d diminished the circulating free fatty acids serving as signaling molecules to trigger the epigenetic regulations of energy metabolism via lipid-sensing transcriptional factors, such as HNF4α. The metabolic disorders induced an unhealthy microenvironment in the metabolically active tissues, ultimately leading to systemic insulin resistance.
Collapse
Affiliation(s)
- Gang Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Li Yang
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shuyue Wang
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Baharan Fekry
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lucas Veillon
- Metabolomic Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Metabolomic Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Berdeaux
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Philip L Lorenzi
- Metabolomic Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cell Biology, MD Anderson Cancer Center-UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
28
|
Erukainure OL, Matsabisa MG, Salau VF, Olofinsan KA, Oyedemi SO, Chukwuma CI, Nde AL, Islam MS. Cannabidiol improves glucose utilization and modulates glucose-induced dysmetabolic activities in isolated rats' peripheral adipose tissues. Biomed Pharmacother 2022; 149:112863. [PMID: 35358799 DOI: 10.1016/j.biopha.2022.112863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/16/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022] Open
Abstract
Reduced glucose uptake and utilization, with concomitant lipolysis in adipose tissues has been linked to the pathogenesis of obesity and its complications. The present study investigated the effect of cannabinoid-stimulated glucose uptake on redox imbalance, glucose and lipid metabolisms, as well as cholinergic and purinergic dysfunctions in isolated rats' adipose tissues. Freshly Isolated rats' adipose tissues were incubated with glucose and different concentrations of cannabidiol for 2 h at 37 °C. The negative control consisted of incubation without cannabidiol, while normal control consisted of incubations without glucose and/or cannabidiol and Metformin served as the standard drug. Cannabidiol caused an increase in adipose-glucose uptake, with concomitant elevation of glutathione, triglyceride level, superoxide dismutase, catalase and 5'nucleoidase activities. It also caused suppression in malondialdehyde and cholesterol levels, acetylcholinesterase, ENTPDase, fructose-1,6-biphosphatase, glucose 6-phosphatase, glycogen phosphorylase, and lipase activities. In silico studies revealed a strong molecular interaction of cannabidiol with adipose triglyceride lipase, hormone-sensitive lipase, and monoglyceride lipase. These results indicate that cannabidiol-enhanced glucose uptake in adipose tissues is associated with enhanced antioxidative activities, concomitant modulation of cholinergic and purinergic dysfunctions, and improved glucose - lipid homeostasis.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Motlalepula G Matsabisa
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| | - Veronica F Salau
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Kolawole A Olofinsan
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban 4000, South Africa
| | - Sunday O Oyedemi
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; Department of Pharmacology, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Chika I Chukwuma
- Center for Quality of Health and Living, Faculty of Health Sciences, Central University of Technology, Bloemfontein 9301, South Africa
| | - Adeline Lum Nde
- Department of Pharmacology, School of Clinical Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, (Westville Campus), Durban 4000, South Africa
| |
Collapse
|
29
|
Reed RM, Nevitt SJ, Kemp GJ, Cuthbertson DJ, Whyte MB, Goff LM. Ectopic fat deposition in populations of black African ancestry: A systematic review and meta-analysis. Acta Diabetol 2022; 59:171-187. [PMID: 34518896 PMCID: PMC8841318 DOI: 10.1007/s00592-021-01797-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
AIMS In populations of black African ancestry (BA), a paradox exists whereby lower visceral adipose tissue is found despite their high risk for type 2 diabetes (T2D). This systematic review investigates ethnic differences in other ectopic fat depots (intrahepatic lipid: IHL; intramyocellular lipid: IMCL and intrapancreatic lipid; IPL) to help contextualise their potential contribution to T2D risk. METHODS A systematic literature search was performed in December 2020 to identify studies reporting at least one ectopic fat comparison between BA and one/more other ethnicity. For IHL, a meta-analysis was carried out with studies considered comparable based on the method of measurement. RESULTS Twenty-eight studies were included (IHL: n = 20; IMCL: n = 8; IPL: n = 4). Meta-analysis of 11 studies investigating IHL revealed that it was lower in BA populations vs pooled ethnic comparators (MD -1.35%, 95% CI -1.55 to -1.16, I2 = 85%, P < 0.00001), white European ancestry (MD -0.94%, 95% CI -1.17 to -0.70, I2 = 79%, P < 0.00001), Hispanic ancestry (MD -2.06%, 95% CI -2.49 to -1.63, I2 = 81%, P < 0.00001) and South Asian ancestry comparators (MD -1.92%, 95% CI -3.26 to -0.57, I2 = 78%, P = 0.005). However, heterogeneity was high in all analyses. Most studies found no significant differences in IMCL between BA and WE. Few studies investigated IPL, however, indicated that IPL is lower in BA compared to WE and HIS. CONCLUSION The discordance between ectopic fat and greater risk for T2D in BA populations raises questions around its contribution to T2D pathophysiology in BA.
Collapse
Affiliation(s)
- Reuben M Reed
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Sarah J Nevitt
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Graham J Kemp
- Department of Musculoskeletal and Ageing Science. Institute of Life Course and Medical Sciences, Liverpool Magnetic Resonance Imaging Centre (LiMRIC), University of Liverpool, Liverpool, UK
| | - Daniel J Cuthbertson
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course Sciences, University of Liverpool, Liverpool, UK
| | - Martin B Whyte
- Faculty of Health & Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Louise M Goff
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| |
Collapse
|
30
|
Qi L, Zushin PJ, Chang CF, Lee YT, Alba DL, Koliwad S, Stahl A. Probing Insulin Sensitivity with Metabolically Competent Human Stem Cell-Derived White Adipose Tissue Microphysiological Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2103157. [PMID: 34761526 PMCID: PMC8776615 DOI: 10.1002/smll.202103157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/21/2021] [Indexed: 05/13/2023]
Abstract
Impaired white adipose tissue (WAT) function has been recognized as a critical early event in obesity-driven disorders, but high buoyancy, fragility, and heterogeneity of primary adipocytes have largely prevented their use in drug discovery efforts highlighting the need for human stem cell-based approaches. Here, human stem cells are utilized to derive metabolically functional 3D adipose tissue (iADIPO) in a microphysiological system (MPS). Surprisingly, previously reported WAT differentiation approaches create insulin resistant WAT ill-suited for type-2 diabetes mellitus drug discovery. Using three independent insulin sensitivity assays, i.e., glucose and fatty acid uptake and suppression of lipolysis, as the functional readouts new differentiation conditions yielding hormonally responsive iADIPO are derived. Through concomitant optimization of an iADIPO-MPS, it is abled to obtain WAT with more unilocular and significantly larger (≈40%) lipid droplets compared to iADIPO in 2D culture, increased insulin responsiveness of glucose uptake (≈2-3 fold), fatty acid uptake (≈3-6 fold), and ≈40% suppressing of stimulated lipolysis giving a dynamic range that is competent to current in vivo and ex vivo models, allowing to identify both insulin sensitizers and desensitizers.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Peter James Zushin
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Ching-Fang Chang
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Yue Tung Lee
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Diana L. Alba
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of California, San Francisco; Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Suneil Koliwad
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of California, San Francisco; Diabetes Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
31
|
Malenica M, Meseldžić N. Oxidative stress and obesity. ARHIV ZA FARMACIJU 2022. [DOI: 10.5937/arhfarm72-36123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Obesity is a disease of excessive accumulation of adipose tissue due to an increased energy intake which is disproportionate to the energy expenditure in the body. The visceral adipose tissue in the obese accumulated in that way increases the risk of developing a number of metabolic and cardiovascular diseases. Disorders such as diabetes, dyslipidemia, inflammation, endothelial dysfunction and mitochondria can contribute to the development of oxidative stress, which is especially pronounced in the abdominal type of obesity. Obesity can induce systemic oxidative stress through a variety of biochemical mechanisms. Although ROS is generated in a large number of cells, mitochondria play a significant role in their intracellular production through the process of oxidative phosphorylation of the respiratory chain, and in fatty acid oxidation reactions. Oxidative stress is a unique link between the various molecular disorders present in the development of insulin resistance that plays a key role in the pathogenesis and progression of chronic metabolic, proinflammatory diseases. The progression of insulin resistance is also affected by inflammation. Both of these can be the cause and the consequence of obesity. The synthesis of the inflammatory mediators is induced by oxidative stress, thus bringing the inflammation and the oxidative stress into a very significant relation. This review aims to highlight recent findings on the role of oxidative stress in the pathogenesis of obesity, with special reference to the mechanisms that explain its occurrence.
Collapse
|
32
|
Adipose Tissue Dysfunctions in Response to an Obesogenic Diet Are Reduced in Mice after Transgenerational Supplementation with Omega 3 Fatty Acids. Metabolites 2021; 11:metabo11120838. [PMID: 34940596 PMCID: PMC8706165 DOI: 10.3390/metabo11120838] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022] Open
Abstract
Obesity is characterized by profound alterations in adipose tissue (AT) biology, leading to whole body metabolic disturbances such as insulin resistance and cardiovascular diseases. These alterations are related to the development of a local inflammation, fibrosis, hypertrophy of adipocytes, and dysregulation in energy homeostasis, notably in visceral adipose tissue (VAT). Omega 3 (n-3) fatty acids (FA) have been described to possess beneficial effects against obesity-related disorders, including in the AT; however, the long-term effect across generations remains unknown. The current study was conducted to identify if supplementation with n-3 polyunsaturated FA (PUFA) for three generations could protect from the consequences of an obesogenic diet in VAT. Young mice from the third generation of a lineage receiving a daily supplementation (1% of the diet) with fish oil rich in eicosapentaenoic acid (EPA) or an isocaloric amount of sunflower oil, were fed a high-fat, high-sugar content diet for 4 months. We explore the transcriptomic adaptations in each lineage using DNA microarray in VAT and bioinformatic exploration of biological regulations using online databases. Transgenerational intake of EPA led to a reduced activation of inflammatory processes, perturbation in metabolic homeostasis, cholesterol metabolism, and mitochondrial functions in response to the obesogenic diet as compared to control mice from a control lineage. This suggests that the continuous intake of long chain n-3 PUFA could be preventive in situations of oversupply of energy-dense, nutrient-poor foods.
Collapse
|
33
|
Chueire VB, Muscelli E. Effect of free fatty acids on insulin secretion, insulin sensitivity and incretin effect - a narrative review. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:24-31. [PMID: 33320449 PMCID: PMC10528699 DOI: 10.20945/2359-3997000000313] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 09/26/2020] [Indexed: 11/23/2022]
Abstract
Deleterious effects of free fatty acids, FFAs, on insulin sensitivity are observed in vivo studies in humans. Mechanisms include impaired insulin signaling, oxidative stress, inflammation, and mitochondrial dysfunction, but the effects on insulin secretion are less well known. Our aim was to review the relationship of increased FFAs with insulin resistance, secretion and mainly with the incretin effect in humans. Narrative review. Increased endogenous or administered FFAs induce insulin resistance. FFAs effects on insulin secretion are debatable; inhibition and stimulation have been reported, depending on the type and duration of lipids exposition and the study subjects. Chronically elevated FFAs seem to decrease insulin biosynthesis, glucose-stimulated insulin secretion and β-cell glucose sensitivity. Lipids infusion decreases the response to incretins with unchanged incretin levels in volunteers with normal glucose tolerance. In contrast, FFAs reduction by acipimox did not restore the incretin effect in type-2 diabetes, probably due to the dysfunctional β-cell. Possible mechanisms of FFAs excess on incretin effect include reduction of the expression and levels of GLP-1 (glucagon like peptide-1) receptor, reduction of connexin-36 expression thus the coordinated secretory activity in response to GLP-1, and GIP (glucose-dependent insulinotropic polypeptide) receptors downregulation in islets cells. Increased circulating FFAs impair insulin sensitivity. Effects on insulin secretion are complex and controversial. Deleterious effects on the incretin-induced potentiation of insulin secretion were reported. More investigation is needed to better understand the extent and mechanisms of β-cell impairment and insulin resistance induced by increased FFAs and how to prevent them.
Collapse
Affiliation(s)
- Valeria Bahdur Chueire
- Departamento de Endocrinologia, Hospital da Pontifícia Universidade Católica de Campinas, Campinas, SP, Brasil,
| | - Elza Muscelli
- Departamento de Clínica Médica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brasil
| |
Collapse
|
34
|
Htun KT, Pan J, Pasanta D, Tungjai M, Udomtanakunchai C, Petcharoen T, Chamta N, Kosicharoen S, Chukua K, Lai C, Kothan S. Advanced Molecular Imaging (MRI/MRS/ 1H NMR) for Metabolic Information in Young Adults with Health Risk Obesity. Life (Basel) 2021; 11:1035. [PMID: 34685406 PMCID: PMC8541404 DOI: 10.3390/life11101035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Obesity or being overweight is a medical condition of abnormal body fat accumulation which is associated with a higher risk of developing metabolic syndrome. The distinct body fat depots on specific parts of the anatomy have unique metabolic properties and different types of regional excessive fat distribution can be a disease hazard. The aim of this study was to identify the metabolome and molecular imaging phenotypes among a young adult population. METHODS The amount and distribution of fat and lipid metabolites profile in the abdomen, liver, and calf muscles of 46 normal weight, 17 overweight, and 13 obese participants were acquired using MRI and MR spectroscopy (MRS), respectively. The serum metabolic profile was obtained using proton NMR spectroscopy. NMR spectra were integrated into seven integration regions, which reflect relative metabolites. RESULTS A significant metabolic disorder symptom appeared in the overweight and obese group, and increased lipid deposition occurred in the abdomen, hepatocytes, and muscles that were statistically significant. Overall, the visceral fat depots had a marked influence on dyslipidemia biomarkers, blood triglyceride (r = 0.592, p < 0.001), and high-density lipoprotein cholesterol (r = -0.484, p < 0.001). Intrahepatocellular lipid was associated with diabetes predictors for hemoglobin (HbA1c%; r = 0.379, p < 0.001) and for fasting blood sugar (r = 0.333, p < 0.05). The lipid signals in serum triglyceride and glucose signals gave similar correspondence to biochemical lipid profiles. CONCLUSIONS This study proves the association between alteration in metabolome in young adults, which is the key population for early prevention of obesity and metabolic syndrome. This study suggests that dyslipidemia prevalence is influenced mainly by the visceral fat depot, and liver fat depot is a key determinant for glucose metabolism and hyperglycemia. Moreover, noninvasive advanced molecular imaging completely elucidated the impact of fat distribution on the anthropometric and laboratory parameters, especially indices of the metabolic syndrome biomarkers in young adults.
Collapse
Affiliation(s)
- Khin Thandar Htun
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Jie Pan
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
- Shandong Provincial Key Laboratory of Animal Resistant Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Duanghathai Pasanta
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Montree Tungjai
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Chatchanok Udomtanakunchai
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Thanaporn Petcharoen
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Nattacha Chamta
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Supak Kosicharoen
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Kiattisak Chukua
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| | - Christopher Lai
- Health and Social Sciences, Singapore Institute of Technology, 10 Dover Drive, Singapore 138683, Singapore;
| | - Suchart Kothan
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (K.T.H.); (D.P.); (M.T.); (C.U.); (T.P.); (N.C.); (S.K.); (K.C.)
| |
Collapse
|
35
|
Morys F, Dadar M, Dagher A. Association Between Midlife Obesity and Its Metabolic Consequences, Cerebrovascular Disease, and Cognitive Decline. J Clin Endocrinol Metab 2021; 106:e4260-e4274. [PMID: 33677592 PMCID: PMC8475210 DOI: 10.1210/clinem/dgab135] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Indexed: 01/08/2023]
Abstract
CONTEXT Chronic obesity is associated with several complications, including cognitive impairment and dementia. However, we have only piecemeal knowledge of the mechanisms linking obesity to central nervous system damage. Among candidate mechanisms are other elements of obesity-associated metabolic syndrome, such as hypertension, dyslipidemia, and diabetes, but also systemic inflammation. While there have been several neuroimaging studies linking adiposity to changes in brain morphometry, a comprehensive investigation of the relationship has so far not been done. OBJECTIVE To identify links between adiposity and cognitive dysfunction. METHODS This observational cohort study (UK Biobank), with an 8-year follow-up, included more than 20 000 participants from the general community, with a mean age of 63 years. Only participants with data available on both baseline and follow-up timepoints were included. The main outcome measures were cognitive performance and mediator variables: hypertension, diabetes, systemic inflammation, dyslipidemia, gray matter measures, and cerebrovascular disease (volume of white matter hyperintensities on magnetic resonance imaging). RESULTS Using structural equation modeling, we found that body mass index, waist-to-hip ratio, and body fat percentage were positively related to higher plasma C-reactive protein, dyslipidemia, hypertension, and diabetes. In turn, hypertension and diabetes were related to cerebrovascular disease. Finally, cerebrovascular disease was associated with lower cortical thickness and volume and higher subcortical volumes, but also cognitive deficits (largest significant pcorrected = 0.02). CONCLUSIONS We show that adiposity is related to poor cognition, with metabolic consequences of obesity and cerebrovascular disease as potential mediators. The outcomes have clinical implications, supporting a role for the management of adiposity in the prevention of late-life dementia and cognitive decline.
Collapse
Affiliation(s)
- Filip Morys
- Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
- Correspondence: Filip Morys, Ph.D., Université McGill, 3801 University Street, H3A 2B4 Montreal, Canada.
| | - Mahsa Dadar
- Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Laval University, Québec, Canada
| | - Alain Dagher
- Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
36
|
Zuccaro A, Zapatería B, Sánchez-Alonso MG, Haro M, Limones M, Terrados G, Izquierdo A, Corrales P, Medina-Gómez G, Herradón G, Sevillano J, Ramos-Álvarez MDP. Pleiotrophin Deficiency Induces Browning of Periovarian Adipose Tissue and Protects against High-Fat Diet-Induced Hepatic Steatosis. Int J Mol Sci 2021; 22:9261. [PMID: 34502170 PMCID: PMC8431550 DOI: 10.3390/ijms22179261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/14/2021] [Accepted: 08/20/2021] [Indexed: 01/14/2023] Open
Abstract
(1) Background: Pleiotrophin preserves insulin sensitivity, regulates adipose tissue lipid turnover and plasticity, energy metabolism and thermogenesis. The aim of this study was to determine the role of pleiotrophin in hepatic lipid metabolism and in the metabolic crosstalk between the liver and brown and white adipose tissue (AT) in a high-fat diet-induced (HFD) obesity mice model. (2) Methods: We analyzed circulating variables, lipid metabolism (hepatic lipid content and mRNA expression), brown AT thermogenesis (UCP-1 expression) and periovarian AT browning (brown adipocyte markers mRNA and immunodetection) in Ptn-/- mice either fed with standard-chow diet or with HFD and in their corresponding Ptn+/+ counterparts. (3) Results: HFD-Ptn-/- mice are protected against the development of HFD-induced insulin resistance, had lower liver lipid content and lower expression of the key enzymes involved in triacylglycerides and fatty acid synthesis in liver. HFD-Ptn-/- mice showed higher UCP-1 expression in brown AT. Moreover, Ptn deletion increased the expression of specific markers of brown/beige adipocytes and was associated with the immunodetection of UCP-1 enriched multilocular adipocytes in periovarian AT. (4) Conclusions: Ptn deletion protects against the development of HFD-induced insulin resistance and liver steatosis, by increasing UCP-1 expression in brown AT and promoting periovarian AT browning.
Collapse
Affiliation(s)
- Agata Zuccaro
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - Begoña Zapatería
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María Gracia Sánchez-Alonso
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María Haro
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María Limones
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - Gloria Terrados
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - Adriana Izquierdo
- Department of Basic Sciences of Health, Facultad Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (A.I.); (P.C.); (G.M.-G.)
| | - Patricia Corrales
- Department of Basic Sciences of Health, Facultad Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (A.I.); (P.C.); (G.M.-G.)
| | - Gema Medina-Gómez
- Department of Basic Sciences of Health, Facultad Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain; (A.I.); (P.C.); (G.M.-G.)
| | - Gonzalo Herradón
- Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain;
| | - Julio Sevillano
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| | - María del Pilar Ramos-Álvarez
- Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28925 Alcorcón, Spain; (A.Z.); (B.Z.); (M.G.S.-A.); (M.H.); (M.L.); (G.T.); (M.d.P.R.-Á.)
| |
Collapse
|
37
|
Hakim O, Bello O, Ladwa M, Shojaee-Moradie F, Jackson N, Peacock JL, Umpleby AM, Charles-Edwards G, Amiel SA, Goff LM. Adiponectin is associated with insulin sensitivity in white European men but not black African men. Diabet Med 2021; 38:e14571. [PMID: 33783876 DOI: 10.1111/dme.14571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/15/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023]
Abstract
AIMS We aimed to assess ethnic differences in inflammatory markers and their relationships with insulin sensitivity and regional adiposity between white European and black African men. METHODS A total of 53 white European and 53 black African men underwent assessment of inflammatory markers alongside Dixon-magnetic resonance imaging to quantify subcutaneous and visceral adipose tissue and intrahepatic lipid. A hyperinsulinaemic-euglycaemic clamp was used to measure whole-body and adipose tissue insulin sensitivity. To assess ethnic differences in relationships, the statistical significance of an interaction term between adipokines and ethnic group was tested in multivariable regression models. RESULTS The black African men exhibited significantly lower adiponectin and tumour necrosis factor-α (TNF-α) and greater interleukin-10 (IL-10) compared to white European men (all p < 0.05). There were no statistically significant ethnic differences in leptin, resistin, IL-6, interferon-γ, IL-13, IL-1β, IL-8 and vascular endothelial growth factor. Several relationships differed significantly by ethnicity such that they were stronger in white European than black African men including IL-6 with visceral adipose tissue; adiponectin with subcutaneous adipose tissue; leptin with intrahepatic lipid; adiponectin, IL-6 and TNF-α with whole-body insulin sensitivity and TNF-α with adipose tissue insulin sensitivity (all pinteraction <0.05). Leptin significantly predicted whole-body insulin sensitivity in white European (R2 = 0.51) and black African (R2 = 0.29) men; however, adiponectin was a statistically significant predictor in only white European men (R2 = 0.22). CONCLUSIONS While adiponectin is lower in black African men, its insulin sensitising effects may be greater in white men suggesting that the role of adipokines in the development of type 2 diabetes may differ by ethnicity.
Collapse
Affiliation(s)
- Olah Hakim
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Oluwatoyosi Bello
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Meera Ladwa
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | | | - Nicola Jackson
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Janet L Peacock
- Department of Epidemiology Geisel, School of Medicine at Dartmouth, Dartmouth, NH, USA
- School of Population Health and Environmental Sciences, King's College London, London, UK
| | - A Margot Umpleby
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Geoffrey Charles-Edwards
- Medical Physics, Guy's and St Thomas, NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Stephanie A Amiel
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Louise M Goff
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
38
|
Furbetta N, Comandatore A, Gianardi D, Palmeri M, Di Franco G, Guadagni S, Caprili G, Bianchini M, Fatucchi LM, Picchi M, Bastiani L, Biancofiore G, Di Candio G, Morelli L. Perioperative Nutritional Aspects in Total Pancreatectomy: A Comprehensive Review of the Literature. Nutrients 2021; 13:1765. [PMID: 34067286 PMCID: PMC8224756 DOI: 10.3390/nu13061765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 02/05/2023] Open
Abstract
Total pancreatectomy (TP) is a highly invasive procedure often performed in patients affected by anorexia, malabsorption, cachexia, and malnutrition, which are risk factors for bad surgical outcome and even may cause enhanced toxicity to chemo-radiotherapy. The role of nutritional therapies and the association between nutritional aspects and the outcome of patients who have undergone TP is described in some studies. The aim of this comprehensive review is to summarize the available recent evidence about the influence of nutritional factors in TP. Preoperative nutritional and metabolic assessment, but also intra-operative and post-operative nutritional therapies and their consequences, are analyzed in order to identify the aspects that can influence the outcome of patients undergoing TP. The results of this review show that preoperative nutritional status, sarcopenia, BMI and serum albumin are prognostic factors both in TP for pancreatic cancer to support chemotherapy, prevent recurrence and prolong survival, and in TP with islet auto-transplantation for chronic pancreatitis to improve postoperative glycemic control and obtain better outcomes. When it is possible, enteral nutrition is always preferable to parenteral nutrition, with the aim to prevent or reduce cachexia. Nowadays, the nutritional consequences of TP, including diabetes control, are improved and become more manageable.
Collapse
Affiliation(s)
- Niccolò Furbetta
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Desirée Gianardi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Matteo Palmeri
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Simone Guadagni
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Giovanni Caprili
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Matteo Bianchini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Lorenzo Maria Fatucchi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Martina Picchi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Luca Bastiani
- Institute of Clinical Physiology, National Council of Research, 56124 Pisa, Italy;
| | | | - Giulio Di Candio
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (N.F.); (A.C.); (D.G.); (M.P.); (G.D.F.); (S.G.); (G.C.); (M.B.); (L.M.F.); (M.P.); (G.D.C.)
| |
Collapse
|
39
|
Dietary potato intake and risks of type 2 diabetes and gestational diabetes mellitus. Clin Nutr 2021; 40:3754-3764. [PMID: 34130021 DOI: 10.1016/j.clnu.2021.04.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND & AIMS Evidence regarding associations between potato consumption and type 2 diabetes (T2D) and gestational diabetes mellitus (GDM) risks is accumulating. This study aims to synthesize the evidence by conducting a meta-analysis of available studies. METHODS PubMed, Web of Science, EMBASE and Cochrane Library were searched (up to August 2020) to retrieve all eligible studies on the associations of interest. The risk estimates with 95% confidence intervals (CIs) were summarized using random- or fixed-effects model based on heterogeneity. Meta-analyses were performed for East and West regions separately. Dose-response relationship was assessed using data from all intake categories in each study. RESULTS A total of 19 studies (13 for T2D; 6 for GDM) were identified, including 21,357 T2D cases among 323,475 participants and 1516 GDM cases among 29,288 pregnancies. Meta-analysis detected a significantly positive association with T2D risk for total potato (RR: 1.19 [1.06, 1.34]), baked/boiled/mashed potato (RR: 1.08 [1.00, 1.16]), and French fries/fried potato (RR: 1.33 [1.03, 1.70]) intakes among Western populations. Dose-response meta-analysis demonstrated a significantly increased T2D risk by 10% (95% CI: 1.07, 1.14; P for trend<0.001), 2% (95% CI: 1.00, 1.04; P for trend = 0.02) and 34% (95% CI: 1.24, 1.46; P for trend<0.001) for each 80 g/day (serving) increment in total potato, unfried potato, and fried potato intakes, respectively. As for GDM, summarized estimates also suggested a higher though non-significant GDM risk for total potato (RR: 1.19 [0.89, 1.58]), and French fries/fried potato (RR: 1.03 [0.97, 1.09]) intakes in Western countries. In the dose-response meta-analysis, a significantly increased GDM risk was revealed for each daily serving (80 g) intakes of total potato (RR: 1.22; 95% CI: 1.06, 1.42; P for trend = 0.007) and unfried potato (RR: 1.26; 95% CI: 1.07, 1.48; P for trend = 0.006). CONCLUSIONS This study suggests that higher potato intake is associated with higher T2D risk among Western populations. The positive relationship presents a significant dose-response manner. Wisely controlled potato consumption may confer potential glucometabolic benefits.
Collapse
|
40
|
IRS-2/Akt/GSK-3 β/Nrf2 Pathway Contributes to the Protective Effects of Chikusetsu Saponin IVa against Lipotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8832318. [PMID: 33884100 PMCID: PMC8041533 DOI: 10.1155/2021/8832318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/23/2021] [Accepted: 03/16/2021] [Indexed: 12/02/2022]
Abstract
Chronic hyperlipidemia leads to pancreatic β-cell apoptosis and dysfunction through inducing oxidative stress. Chikusetsu saponin IVa (CHS) showed antioxidant and antidiabetic properties in our previous studies; however, its protective effects against lipotoxicity-induced β-cell oxidative stress and dysfunction are not clear. This study was designed to investigate the effects of CHS against lipotoxicity-induced β-cell injuries and its possible mechanism involved. High-fat (HF) diet and a low dose of streptozotocin- (STZ-) induced type 2 diabetes mellitus (T2DM) model in vivo and βTC3 cells subjected to 0.5 mM palmitate (PA) to imitate the lipotoxic model in vitro were performed. Pancreatic functions, ROS, and antioxidant protein measurements were performed to evaluate the effects of CHS on cell injuries. Protein expression levels were measured by Western blotting. Furthermore, siRNA-targeted Nrf2, PI3K/Akt inhibitor (LY294002), or GSK-3β inhibitor (LiCl) was used to investigate the crosstalk relationships between proteins. As the results showed, CHS treatment inhibited apoptosis, promoted insulin release, and reduced oxidative stress. CHS treatment significantly increased the expression of Nrf2 in the cytoplasm and nuclear protein. The antioxidative and benefit effects of CHS were inhibited by siNrf2. The phosphorylation of IRS-2, PI3K, Akt, and GSK-3β was markedly increased by CHS which were inhibited by PA. In addition, inhibition of PI3K/Akt or GSK-3β with specific inhibitors dramatically abrogated the protective effects of CHS, revealing that the IRS-2/Akt/GSK-3β signaling axis was involved in the protective effects of CHS. These results demonstrate that CHS protected βTC3 cells against PA-induced oxidative stress and cell dysfunction through Nrf2 by the IRS-2/Akt/GSK-3β-mediated pathway.
Collapse
|
41
|
Blair HR, Tomas C, Miwa S, Heath A, Russell A, Ginkel MV, Gunn D, Walker M. Peroxisomes and pancreatic beta-cell lipo-dysfunction. J Diabetes Complications 2021; 35:107843. [PMID: 33419633 DOI: 10.1016/j.jdiacomp.2020.107843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
AIMS Pancreatic beta-cell lipo-dysfunction decreases insulin secretion and predisposes to the development of type 2 diabetes. Through targeted Pex11β knockdown and peroxisome depletion, our aim was to investigate the specific contribution of peroxisomes to palmitate mediated pancreatic beta-cell dysfunction. METHODS MIN6 cells were transfected with probes targeted against Pex11β, a regulator of peroxisome abundance, or with scrambled control probes. Peroxisome abundance was measured by PMP-70 protein expression. 48 h post transfection, cells were incubated with 250 μM palmitate or BSA control for a further 48 h before measurement of glucose stimulated insulin secretion and of reactive oxygen species. RESULTS Pex11β knockdown decreased target gene expression by >80% compared with the scrambled control (P<0.001). This led to decreased PMP-70 expression (p<0.01) and a 22% decrease in peroxisome number (p<0.05). At 25 mM glucose, palmitate treatment decreased insulin secretion by 64% in the scrambled control cells (2.54±0.25 vs 7.07±0.83 [mean±SEM] ng/h/μg protein; Palmitate vs BSA P<0.001), but by just 37% in the Pex11β knockdown cells. Comparing responses in the presence of palmitate, insulin secretion at 25 mM glucose was significantly greater in the Pex11β knockdown cells compared with the scrambled controls (4.04±0.46 vs 2.54±0.25 ng/h/μg protein; p<0.05). Reactive oxygen species generation with palmitate was lower in the Pex11β knockdown cells compared with the scrambled controls (P<0.001). CONCLUSION Pex11β knockdown decreased peroxisome abundance, decreased palmitate mediated reactive oxygen species generation, and reversed the inhibitory effect of palmitate on insulin secretion. These findings reveal a distinct role of peroxisomes in palmitate mediated beta-cell dysfunction.
Collapse
Affiliation(s)
- Helen R Blair
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Cara Tomas
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Alan Heath
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Alison Russell
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | | | - David Gunn
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Mark Walker
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
42
|
Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, Hu J, Fleming I, Wang DW. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther 2021; 6:94. [PMID: 33637672 PMCID: PMC7910446 DOI: 10.1038/s41392-020-00443-w] [Citation(s) in RCA: 595] [Impact Index Per Article: 148.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 01/31/2023] Open
Abstract
The arachidonic acid (AA) pathway plays a key role in cardiovascular biology, carcinogenesis, and many inflammatory diseases, such as asthma, arthritis, etc. Esterified AA on the inner surface of the cell membrane is hydrolyzed to its free form by phospholipase A2 (PLA2), which is in turn further metabolized by cyclooxygenases (COXs) and lipoxygenases (LOXs) and cytochrome P450 (CYP) enzymes to a spectrum of bioactive mediators that includes prostanoids, leukotrienes (LTs), epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid (diHETEs), eicosatetraenoic acids (ETEs), and lipoxins (LXs). Many of the latter mediators are considered to be novel preventive and therapeutic targets for cardiovascular diseases (CVD), cancers, and inflammatory diseases. This review sets out to summarize the physiological and pathophysiological importance of the AA metabolizing pathways and outline the molecular mechanisms underlying the actions of AA related to its three main metabolic pathways in CVD and cancer progression will provide valuable insight for developing new therapeutic drugs for CVD and anti-cancer agents such as inhibitors of EETs or 2J2. Thus, we herein present a synopsis of AA metabolism in human health, cardiovascular and cancer biology, and the signaling pathways involved in these processes. To explore the role of the AA metabolism and potential therapies, we also introduce the current newly clinical studies targeting AA metabolisms in the different disease conditions.
Collapse
Affiliation(s)
- Bei Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Lujin Wu
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jing Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China.
| |
Collapse
|
43
|
Boldarine VT, Joyce E, Pedroso AP, Telles MM, Oyama LM, Bueno AA, Ribeiro EB. Oestrogen replacement fails to fully revert ovariectomy-induced changes in adipose tissue monoglycerides, diglycerides and cholesteryl esters of rats fed a lard-enriched diet. Sci Rep 2021; 11:3841. [PMID: 33589704 PMCID: PMC7884784 DOI: 10.1038/s41598-021-82837-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/22/2021] [Indexed: 01/09/2023] Open
Abstract
Menopause may be accompanied by abdominal obesity and inflammation, conditions accentuated by high-fat intake, especially of saturated fat (SFA)-rich diets. We investigated the consequences of high-SFA intake on the fatty acid (FA) profile of monoglycerides, diglycerides and cholesteryl esters from retroperitoneal white adipose tissue (RET) of rats with ovariectomy-induced menopause, and the effect of oestradiol replacement. Wistar rats were either ovariectomized (Ovx) or sham operated (Sham) and fed either standard chow (C) or lard-enriched diet (L) for 12 weeks. Half of the Ovx rats received 17β-oestradiol replacement (Ovx + E2). Body weight and food intake were measured weekly. RET neutral lipids were chromatographically separated and FAs analysed by gas chromatography. Ovariectomy alone increased body weight, feed efficiency, RET mass, leptin and insulin levels, leptin/adiponectin ratio, HOMA-IR and HOMA-β indexes. OvxC + E2 showed attenuation in nearly all blood markers. HOMA-β index was restored in OvxL + E2. OvxC showed significantly disturbed SFA and polyunsaturated FA (PUFA) profile in RET cholesteryl esters (CE). OvxC also showed increased monounsaturated FA (MUFA) in the monoglyceride diglyceride (Mono-Di) fraction. Similar changes were not observed in OvxL, although increased SFA and decreased PUFA was observed in Mono-Di. Overall, HRT was only partially able to revert changes induced by ovariectomy. There appears to be increased mobilization of essential FA in Ovx via CE, which is a dynamic lipid species. The same results were not found in Mono-Di, which are more inert. HRT may be helpful to preserve FA profile in visceral fat, but possibly not wholly sufficient in reverting the metabolic effects induced by menopause.
Collapse
Affiliation(s)
- Valter Tadeu Boldarine
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil.
| | - Ellen Joyce
- Department of Biological Sciences, College of Health, Life and Environmental Sciences, University of Worcester, Worcester, UK
| | - Amanda Paula Pedroso
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| | - Mônica Marques Telles
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| | - Lila Missae Oyama
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| | - Allain Amador Bueno
- Department of Biological Sciences, College of Health, Life and Environmental Sciences, University of Worcester, Worcester, UK
| | - Eliane Beraldi Ribeiro
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| |
Collapse
|
44
|
Buch C, Muller T, Leemput J, Passilly-Degrace P, Ortega-Deballon P, Pais de Barros JP, Vergès B, Jourdan T, Demizieux L, Degrace P. Endocannabinoids Produced by White Adipose Tissue Modulate Lipolysis in Lean but Not in Obese Rodent and Human. Front Endocrinol (Lausanne) 2021; 12:716431. [PMID: 34434170 PMCID: PMC8382141 DOI: 10.3389/fendo.2021.716431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
White adipose tissue (WAT) possesses the endocannabinoid system (ECS) machinery and produces the two major endocannabinoids (ECs), arachidonoylethanolamide (AEA) and 2-arachidonoylglycerol (2-AG). Accumulating evidence indicates that WAT cannabinoid 1 receptors (CB1R) are involved in the regulation of fat storage, tissue remodeling and secretory functions but their role in controlling lipid mobilization is unclear. In the present study, we used different strategies to acutely increase ECS activity in WAT and tested the consequences on glycerol production as a marker of lipolysis. Treating lean mice or rat WAT explants with JLZ195, which inhibits ECs degrading enzymes, induced an increase in 2-AG tissue contents that was associated with a CB1R-dependent decrease in lipolysis. Direct treatment of rat WAT explants with AEA also inhibited glycerol production while mechanistic studies revealed it could result from the stimulation of Akt-signaling pathway. Interestingly, AEA treatment decreased lipolysis both in visceral and subcutaneous WAT collected on lean subjects suggesting that ECS also reduces fat store mobilization in Human. In obese mice, WAT content and secretion rate of ECs were higher than in control while glycerol production was reduced suggesting that over-produced ECs may inhibit lipolysis activating local CB1R. Strikingly, our data also reveal that acute CB1R blockade with Rimonabant did not modify lipolysis in vitro in obese mice and human explants nor in vivo in obese mice. Taken together, these data provide physiological evidence that activation of ECS in WAT, by limiting fat mobilization, may participate in the progressive tissue remodeling that could finally lead to organ dysfunction. The present findings also indicate that acute CB1R blockade is inefficient in regulating lipolysis in obese WAT and raise the possibility of an alteration of CB1R signaling in conditions of obesity.
Collapse
Affiliation(s)
- Chloé Buch
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Tania Muller
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Julia Leemput
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Patricia Passilly-Degrace
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pablo Ortega-Deballon
- Department of Digestive, Thoracic and Surgical Oncology, University Hospital, Dijon, France
| | | | - Bruno Vergès
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
- Department of Endocrinology-Diabetology, University Hospital, Dijon, France
| | - Tony Jourdan
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Laurent Demizieux
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pascal Degrace
- Team Pathophysiology of Dyslipidemia, INSERM UMR1231, Université de Bourgogne Franche-Comté, Dijon, France
- *Correspondence: Pascal Degrace,
| |
Collapse
|
45
|
Hu HQ, Qiao JT, Liu FQ, Wang JB, Sha S, He Q, Cui C, Song J, Zang N, Wang LS, Sun Z, Chen L, Hou XG. The STING-IRF3 pathway is involved in lipotoxic injury of pancreatic β cells in type 2 diabetes. Mol Cell Endocrinol 2020; 518:110890. [PMID: 32781250 DOI: 10.1016/j.mce.2020.110890] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
Lipotoxic injury of pancreatic β cells is an important pathological feature in type 2 diabetes mellitus (T2DM). Stimulator of interferon genes (STING) can recognize its own DNA leaked into the cytoplasm from damaged mitochondria or nuclei of the host cell, thus activating its downstream factor interferon regulatory factor 3 (IRF3), causing inflammation and apoptosis. The STING-IRF3 signaling pathway is closely related to glycolipid metabolism, but its relationship with the lipotoxicity of pancreatic β cells has rarely been reported. Here, we investigated the role of the STING-IRF3 signaling pathway in lipotoxicity-induced inflammation, apoptosis, and dysfunction of pancreatic β cells. We examined the activation of STING and IRF3 in islets of db/db mice and identified the role of the STING-IRF3 signaling pathway in palmitic acid (PA)-induced lipotoxic injury of INS-1, a rat insulinoma cell line. STING and phosphorylated IRF3 including downstream interferon-β were upregulated in islets of db/db mice and PA-induced INS-1 cells. Gene silencing of STING or IRF3 ameliorated PA-induced INS-1 cell inflammation and apoptosis, and reversed impaired insulin synthesis. Additionally, PA induced downregulation of the phosphoinositide 3-kinase-AKT signaling pathway, and impaired high glucose-stimulated insulin secretion was reversed after knockdown of STING or IRF3. Our results suggest that activation of the STING-IRF3 pathway triggers inflammation and apoptosis of pancreatic β cells, leading to β-cell damage and dysfunction. Hence, inhibition of this signaling pathway may represent a novel approach for β-cell protection in T2DM.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cells, Cultured
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/physiology
- Interferon Regulatory Factor-3/physiology
- Male
- Membrane Proteins/physiology
- Mice
- Mice, Transgenic
- Palmitic Acid/pharmacology
- Palmitic Acid/toxicity
- Phosphatidylinositol 3-Kinases/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- H Q Hu
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - J T Qiao
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - F Q Liu
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan 250012, China
| | - J B Wang
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - S Sha
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - Q He
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - C Cui
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - J Song
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - N Zang
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - L S Wang
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China
| | - Z Sun
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - L Chen
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan 250012, China.
| | - X G Hou
- Department of Endocrine and Metabolism, Qilu Hospital of Shandong University, Jinan, Shandong, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, Shandong, China; Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan 250012, China.
| |
Collapse
|
46
|
Jebril M, Liu X, Shi Z, Mazidi M, Altaher A, Wang Y. Prevalence of Type 2 Diabetes and Its Association with Added Sugar Intake in Citizens and Refugees Aged 40 or Older in the Gaza Strip, Palestine. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E8594. [PMID: 33228087 PMCID: PMC7699327 DOI: 10.3390/ijerph17228594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Little is known about the prevalence and risk factors of diabetes among Gaza Palestinians, 64% of whom are refugees with exceeded sugar intake. We aimed to estimate the prevalence of type 2 diabetes (T2D) and its association with added sugar intake among residents, with regular visits to primary healthcare centers (PHCs) across Gaza. METHODS From October to December of 2019, a cross-sectional survey was conducted among 1000 citizens and refugees in nine PHCs selected from the five governorates of the Gaza Strip. Information on dietary intake, medical history, and other risk factors was collected by trained health workers, using structured questionnaires. Anthropometry and biochemical data were extracted from the PHC medical record system. RESULTS Overall, the prevalence of diagnosed T2D and undiagnosed T2D were 45.2% and 16.8%, respectively, in adults aged 42 to 74 years, with the differences among citizens and refugees (diagnosed: 46.2% vs. 43.8%; undiagnosed: 15.7% vs. 18.2%). The uncontrolled glycaemic rate was 41.9% and 36.8% for diagnosed patients in citizens and refugees, respectively. Among those without a clinical diagnosis of T2D, after multivariable adjustment, daily added sugar intake was positively associated with fasting glucose and the risk of undiagnosed T2D (odds ratio, 95% CI, highest vs. lowest intake, was 2.71 (1.12-6.54) (pfor trend < 0.001). In stratified analysis, the associations between added sugar intake and the risk of undiagnosed T2D tend to be stronger among refugees or those with higher body mass index. CONCLUSIONS Among Palestinian adults, both citizens and refugees are affected by T2D. Added sugar intake is associated with the risk of undiagnosed T2D.
Collapse
Affiliation(s)
- Majed Jebril
- Global Health Institute, School of Public Health, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an 710061, China;
| | - Xin Liu
- Global Health Institute, School of Public Health, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an 710061, China;
| | - Zumin Shi
- Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Mohsen Mazidi
- Department of Twin Research and Genetic Epidemiology, King’s College London, St Thomas’ Hospital, Strand, London SE1 7EH, UK;
| | - Akram Altaher
- Department of Medical Sciences, University College of Science & Technology, Khan Younis 950, Palestine;
| | - Youfa Wang
- Global Health Institute, School of Public Health, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an 710061, China;
| |
Collapse
|
47
|
Erukainure OL, Salau VF, Bharuth V, Koorbanally NA, Islam MS. Hyperglycemia alters lipid metabolism and ultrastructural morphology of cerebellum in brains of diabetic rats: Therapeutic potential of raffia palm (Raphia hookeri G. Mann & H. Wendl) wine. Neurochem Int 2020; 140:104849. [PMID: 32927025 DOI: 10.1016/j.neuint.2020.104849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
The present study investigated the effect of raffia palm (Raphia hookeri) wine (RPW) on hyperglycemia-mediated lipid metabolites and pathways, functional chemistry and ultrastructural morphology of cerebellums in type 2 diabetes (T2D). T2D was induced in male Sprague-Dawley rats by feeding with 10% fructose ad libitum for 2 weeks before injecting intraperitoneally with 40 mg/kg bodyweight (bw) streptozotocin. Following confirmation of hyperglycemia at blood glucose >200 mg/dL, diabetic rats were treated with RPW at 150 and 300 mg/kg bw respectively. Metformin served as the standard drug. Negative and normal controls consisted of untreated diabetic and non-diabetic rats, respectively. After 5 weeks of treatment, the rats were humanely sacrificed, and their cerebellum excised from the harvested brains. GC-MS analysis revealed significant alterations in cerebellar lipid metabolites depicted by changes in unsaturated and saturated fatty acids, fatty - esters, alcohols, and amides, glycols and steroids on induction of T2D. Pathway enrichment analysis of the lipid metabolites revealed inactivation of arachidonic metabolic pathway following T2D induction. Treatment with both doses of RPW restored most of the metabolites, while reactivating arachidonic acid metabolism (high dose only). Low dose of RPW led to the activation of retinol metabolism. Both doses of RPW maintained cerebellar functional chemistry as revealed by FTIR analysis. TEM analysis revealed swollen mitochondria, depleted numbers of synaptic vesicles, and shrunk synaptic clefts following induction of T2D. These ultrastructural morphologies were improved in RPW-treated rats. These results portray the therapeutic potential of raffia palm wine in the management of neurodegenerative complications in T2D.
Collapse
Affiliation(s)
- Ochuko L Erukainure
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa; Department of Pharmacology, University of the Free State, Bloemfontein, 9300, South Africa
| | - Veronica F Salau
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Vishal Bharuth
- Microscopy and Microanalysis Unit, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Neil A Koorbanally
- School of Chemistry and Physics, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa.
| |
Collapse
|
48
|
Genders AJ, Holloway GP, Bishop DJ. Are Alterations in Skeletal Muscle Mitochondria a Cause or Consequence of Insulin Resistance? Int J Mol Sci 2020; 21:ijms21186948. [PMID: 32971810 PMCID: PMC7554894 DOI: 10.3390/ijms21186948] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
As a major site of glucose uptake following a meal, skeletal muscle has an important role in whole-body glucose metabolism. Evidence in humans and animal models of insulin resistance and type 2 diabetes suggests that alterations in mitochondrial characteristics accompany the development of skeletal muscle insulin resistance. However, it is unclear whether changes in mitochondrial content, respiratory function, or substrate oxidation are central to the development of insulin resistance or occur in response to insulin resistance. Thus, this review will aim to evaluate the apparent conflicting information placing mitochondria as a key organelle in the development of insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Amanda J. Genders
- Institute for Health and Sport (iHeS), Victoria University, Melbourne 8001, Australia;
- Correspondence: ; Tel.: +61-3-9919-9556
| | - Graham P. Holloway
- Dept. Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - David J. Bishop
- Institute for Health and Sport (iHeS), Victoria University, Melbourne 8001, Australia;
| |
Collapse
|
49
|
Anisonyan AV, Sandler YG, Khaimenova TY, Keyan VA, Saliev KG, Sbikina ES, Vinnitskaya EV. [Non-alcoholic fatty liver disease and type 2 diabetes mellitus: issues of the liver fibrosis diagnostics]. TERAPEVT ARKH 2020; 92:73-78. [PMID: 33346465 DOI: 10.26442/00403660.2020.08.000770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 12/20/2022]
Abstract
AIM To evaluate the frequency of liver fibrosis progression to stage 34 among patients with non-alcoholic fatty liver disease (NAFLD), type 2 diabetes and obesity, to identify predictors of severe liver fibrosis, to propose an algorithm for diagnosing fibrosis in this category of patients. MATERIALS AND METHODS 160 patients with NAFLD, type 2 diabetes mellitus (DM) and obesity and 50 patients with NAFLD without diabetes were comprehensively examined. Patients underwent laboratory examination (clinical blood test, biochemical analysis, immunoglobulins G, M, autoantibody assay, coagulogram), liver ultrasound. All patients underwent determination of the liver fibrosis stage by two methods: the serological test FibroMax and indirect ultrasound elastometry of the liver; 40 patients underwent a liver biopsy. Statistical data processing was performed using the programming language and statistical calculations R: we used correlation analysis, multiple logistic regression method, one-way analysis of variance, multi-factor analysis, the Kruskal-Wallis method, and comparison of the number of patients using the Fisher test. RESULTS DM is a risk factor for the liver fibrosis progression in patients with NAFLD. Significant markers of severe fibrosis in this category of patients are increased levels of GGTP, haptoglobin and alpha-2-macroglobulin, lower platelet and prothrombin levels. Obesity and isolated steatosis without steatohepatitis are not markers of severe liver fibrosis at present, but obesity can be considered a risk factor for the progression of fibrosis in the future. CONCLUSION All patients with NAFLD in combination with diabetes need screening to detect advanced liver fibrosis: it is advisable to determine the levels of GGTP, haptoglobin and alpha-2-macroglobulin.
Collapse
Affiliation(s)
- A V Anisonyan
- Loginov Moscow Clinical Scientific and Practical Center
| | - Y G Sandler
- Loginov Moscow Clinical Scientific and Practical Center
| | | | - V A Keyan
- Loginov Moscow Clinical Scientific and Practical Center
| | - K G Saliev
- Loginov Moscow Clinical Scientific and Practical Center
| | - E S Sbikina
- Loginov Moscow Clinical Scientific and Practical Center
| | | |
Collapse
|
50
|
Petrick HL, Foley KP, Zlitni S, Brunetta HS, Paglialunga S, Miotto PM, Politis-Barber V, O’Dwyer C, Philbrick DJ, Fullerton MD, Schertzer JD, Holloway GP. Adipose Tissue Inflammation Is Directly Linked to Obesity-Induced Insulin Resistance, while Gut Dysbiosis and Mitochondrial Dysfunction Are Not Required. FUNCTION (OXFORD, ENGLAND) 2020; 1:zqaa013. [PMID: 34278304 PMCID: PMC8276887 DOI: 10.1093/function/zqaa013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 01/06/2023]
Abstract
Obesity is associated with adipose tissue hypertrophy, systemic inflammation, mitochondrial dysfunction, and intestinal dysbiosis. Rodent models of high-fat diet (HFD)-feeding or genetic deletion of multifunctional proteins involved in immunity and metabolism are often used to probe the etiology of obesity; however, these models make it difficult to divorce the effects of obesity, diet composition, or immunity on endocrine regulation of blood glucose. We, therefore, investigated the importance of adipose inflammation, mitochondrial dysfunction, and gut dysbiosis for obesity-induced insulin resistance using a spontaneously obese mouse model. We examined metabolic changes in skeletal muscle, adipose tissue, liver, the intestinal microbiome, and whole-body glucose control in spontaneously hyperphagic C57Bl/6J mice compared to lean littermates. A separate subset of lean and obese mice was subject to 8 weeks of obesogenic HFD feeding, or to pair feeding of a standard rodent diet. Hyperphagia, obesity, adipose inflammation, and insulin resistance were present in obese mice despite consuming a standard rodent diet, and these effects were blunted with caloric restriction. However, hyperphagic obese mice had normal mitochondrial respiratory function in all tissues tested and no discernable intestinal dysbiosis relative to lean littermates. In contrast, feeding mice an obesogenic HFD altered the composition of the gut microbiome, impaired skeletal muscle mitochondrial bioenergetics, and promoted poor glucose control. These data show that adipose inflammation and redox stress occurred in all models of obesity, but gut dysbiosis and mitochondrial respiratory dysfunction are not always required for obesity-induced insulin resistance. Rather, changes in the intestinal microbiome and mitochondrial bioenergetics may reflect physiological consequences of HFD feeding.
Collapse
Affiliation(s)
- Heather L Petrick
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Kevin P Foley
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Soumaya Zlitni
- Departments of Genetics and Medicine, Stanford University, Stanford, 94305, CA, USA
| | - Henver S Brunetta
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada,Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Sabina Paglialunga
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Paula M Miotto
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Valerie Politis-Barber
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Conor O’Dwyer
- Department of Biochemistry, Microbiology and Immunology, Centre for Inflammation, Infection and Immunity, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Diana J Philbrick
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Morgan D Fullerton
- Department of Biochemistry, Microbiology and Immunology, Centre for Inflammation, Infection and Immunity, Centre for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada,Address correspondence to G.P.H. (e-mail: )
| |
Collapse
|