1
|
Liu Z, Li Q, Zhao F, Chen J. A decade review on phytochemistry and pharmacological activities of Cynomorium songaricum Rupr.: Insights into metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156602. [PMID: 40058318 DOI: 10.1016/j.phymed.2025.156602] [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: 01/05/2025] [Revised: 02/12/2025] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Cynomorium songaricum Rupr. (CSR), a perennial herb with a rich history in traditional medicine, has demonstrated therapeutic potential against metabolic syndrome (MetS) through its active compounds, including proanthocyanidins, polysaccharides, and triterpenoids. MetS, a global health concern, encompasses interlinked conditions such as obesity, type 2 diabetes mellitus (T2DM), and inflammation. This review synthesizes recent findings on CSR's pharmacological and phytochemical properties, focusing on its role in ameliorating MetS. METHODS Following Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines, relevant studies were retrieved from PubMed, Web of Science, and CNKI databases up to December 2024. Keywords included "Cynomorium Songaricum Rupr.", "Cynomorii Herba", "Suoyang", "Suo Yang", "Metabolic syndrome", "Proanthocyanidins", "Polysaccharides" and "Triterpenoids" and their combinations. Inclusion criteria emphasized studies exploring CSR's impact on MetS, while duplicate, low-quality studies and studies not written in Chinese, English, or unrelated were excluded. RESULTS A total of 92 studies were analyzed, revealing that CSR's active components exhibit multi-target effects. Proanthocyanidins reduce glucose absorption and oxidative stress, polysaccharides enhance insulin sensitivity and gut microbiota composition, and triterpenoids mitigate obesity and mitochondria damage. These mechanisms collectively contribute to the beneficial effects of CSR against MetS. CONCLUSION CSR presents a promising natural therapy for MetS, utilizing its pharmacologically active compounds to address core metabolic dysfunctions. Future studies should focus on clinical validation and safety assessments to facilitate CSR's integration into modern therapeutic regimens.
Collapse
Affiliation(s)
- Zhihao Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, China
| | - Qihao Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Fu Zhao
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Jihang Chen
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China; The Chinese University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D Center, Shenzhen, China.
| |
Collapse
|
2
|
Chen G, Chen Y, Yao Y, Ding L, Wu S, Wu W. High Cumulative Non-High-Density Lipoprotein-Cholesterol Concentration Increases the Risk of New-Onset Arterial Stiffness - A Prospective Cohort Study. Circ J 2025; 89:629-637. [PMID: 40090733 DOI: 10.1253/circj.cj-24-0921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
BACKGROUND The relationship between cumulative non-high-density lipoprotein-cholesterol (cum-non-HDL-C) and the risk of new-onset arterial stiffness has not been characterized. METHODS AND RESULTS A total of 6,852 participants with 3 consecutive measurements of total cholesterol and HDL-C and a baseline brachial-ankle pulse wave velocity (baPWV) <1,400 cm/s during 2010-2011, 2012-2013, and 2014-2015 were included. The cum-non-HDL-C concentrations were determined using time weighting, and the participants were grouped: G1 <130 mg/dL, G2 130-159 mg/dL, G3 160-189 mg/dL, and G4 ≥190 mg/dL. Cox models were used to characterize the relationships between cum-non-HDL-C and arterial stiffness by calculating hazard ratios (HRs) and 95% confidence intervals (CIs). Arterial stiffness (baPWV ≥1,800 cm/s) was present in 327 (4.77%) participants over a median follow-up period of 7.7 (interquartile range 7.2-8.2) years. After adjustment for multiple confounders, G2-4 had adjusted HRs (95% CIs) of 1.12 (0.85, 1.48), 1.45 (1.05, 1.99), and 2.52 (1.69, 3.74), respectively (P=0.0004), vs. G1. The adjusted HRs (95% CIs) for exposures of 2, 4, and 6 years were 1.17 (0.87, 1.58), 1.46 (1.96, 2.01), and 1.67 (1.14, 2.44), respectively (P=0.0029), vs. 0 years. Restricted cubic spline analysis revealed a linear dose-response relationship between cum-non-HDL-C and arterial stiffness risk. CONCLUSIONS A high cum-non-HDL-C concentration and prolonged exposure to this increase the risk of arterial stiffness. The monitoring and maintenance of appropriate cum-non-HDL-C may reduce the risk of arterial stiffness.
Collapse
Affiliation(s)
- Guanzhi Chen
- Peking Union Medical College, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Fuwai Hospital
| | - Yanjuan Chen
- Department of Endocrinology, Second Affiliated Hospital of Shantou University Medical College
| | - Yan Yao
- Peking Union Medical College, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Fuwai Hospital
| | - Ligang Ding
- Peking Union Medical College, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Fuwai Hospital
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital
| | - Weiqiang Wu
- Department of Cardiology, Second Affiliated Hospital of Shantou University Medical College
| |
Collapse
|
3
|
Varghese M, Thekkelnaycke R, Soni T, Zhang J, Maddipati K, Singer K. Sex differences in the lipid profiles of visceral adipose tissue with obesity and gonadectomy. J Lipid Res 2025; 66:100803. [PMID: 40245983 DOI: 10.1016/j.jlr.2025.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/05/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
In obesity, adipose tissue (AT) expansion is accompanied by chronic inflammation. Altered lipid composition in the visceral or gonadal white AT (GWAT) directly drive AT macrophage accumulation and activation to a proinflammatory phenotype. Sex steroid hormones modulate visceral versus subcutaneous lipid accumulation that correlates with metabolic syndrome, especially in men and postmenopausal women who are more prone to abdominal obesity. Prior studies demonstrated sex differences in GWAT lipid species in HFD-fed mice, but the role of sex hormones is still unclear. We hypothesized that sex hormone alterations with gonadectomy (GX) would further impact lipid composition in the obese GWAT. Untargeted lipidomics of obese GWAT identified sex differences in phospholipids, sphingolipids, sterols, fatty acyls, saccharolipids and prenol lipids. Males had significantly more precursor fatty acids (palmitic, oleic, linoleic, and arachidonic acid) than females and GX mice. Targeted lipidomics for fatty acids and oxylipins in the HFD-fed male and female GWAT stromal vascular fraction identified higher omega-6 to omega-3 free fatty acid profile in males and differences in PUFAs-derived prostaglandins, thromboxanes, and leukotrienes. Both obese male and female GWAT stromal vascular fraction showed increased levels of arachidonic acid-derived oxylipins compared to their lean counterparts. Bulk RNA-seq of sorted GWAT AT macrophages highlighted sex and diet differences in PUFA and oxylipin metabolism genes. These findings of sexual dimorphism in both stored lipid species and PUFA-derived mediators with diet and GX emphasize sex differences in lipid metabolism pathways that drive inflammation responses and metabolic disease risk in obesity.
Collapse
Affiliation(s)
- Mita Varghese
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rajendiran Thekkelnaycke
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - Tanu Soni
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | - Jiayu Zhang
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, MI, USA
| | | | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Li G, Meex RCR, Goossens GH. The role of tissue oxygenation in obesity-related cardiometabolic complications. Rev Endocr Metab Disord 2025; 26:19-30. [PMID: 39298040 PMCID: PMC11790814 DOI: 10.1007/s11154-024-09910-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Obesity is a complex, multifactorial, chronic disease that acts as a gateway to a range of other diseases. Evidence from recent studies suggests that changes in oxygen availability in the microenvironment of metabolic organs may exert an important role in the development of obesity-related cardiometabolic complications. In this review, we will first discuss results from observational and controlled laboratory studies that examined the relationship between reduced oxygen availability and obesity-related metabolic derangements. Next, the effects of alterations in oxygen partial pressure (pO2) in the adipose tissue, skeletal muscle and the liver microenvironment on physiological processes in these key metabolic organs will be addressed, and how this might relate to cardiometabolic complications. Since many obesity-related chronic diseases, including type 2 diabetes mellitus, cardiovascular diseases, chronic kidney disease, chronic obstructive pulmonary disease and obstructive sleep apnea, are characterized by changes in pO2 in the tissue microenvironment, a better understanding of the metabolic impact of altered tissue oxygenation can provide valuable insights into the complex interplay between environmental and biological factors involved in the pathophysiology of metabolic impairments. This may ultimately contribute to the development of novel strategies to prevent and treat obesity-related cardiometabolic diseases.
Collapse
Affiliation(s)
- Geng Li
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Ruth C R Meex
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands.
| |
Collapse
|
5
|
Ding YY, Lan J, Wang Y, Pan Y, Song T, Liu S, Gu Z, Ge Y. Structure characterization of Grifola frondosa polysaccharide and its effect on insulin resistance in HFD-fed mice. NPJ Sci Food 2025; 9:3. [PMID: 39774946 PMCID: PMC11707143 DOI: 10.1038/s41538-024-00359-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Polysaccharide extracted from Grifola frondosa (GFP) was selected in this study. After preliminary separation, four factions were collected, named GFP-F1, GFP-F2, GFP-F3 and GFP-F4. GPF-F2 was further separated into two fractions, namely GFP-N1 and GFP-N2. The molecular weight of GFP-N1 and GFP-N2 was 3.323×103 kDa and 10.8 kDa, respectively. GFP-N1 was composed of glucose and galactose and 1 → 3, 1 → 4, and 1 → 6 glycosidic bonds. GFP-N2 was composed of glucose, galactose and mannose and 1 → 2, 1 → 3, 1 → 4, and 1 → 6 glycosidic bonds. GFP could significantly relieve the insulin resistance induced by HFD. GFP significantly alleviated gut microbiota disturbance caused by HFD and increased the production of short-chain fatty acids, and further reduced the expression of LPS/TLR4 inflammatory pathway. GFP significantly reduced the oxidative stress induced by HFD, increased the expression of the Nrf2/ARE signaling pathway. These results indicated that GFP could be developed as a potential ingredient for the management of insulin resistance.
Collapse
Affiliation(s)
- Yin-Yi Ding
- National Experimental Teaching Demonstration Center of Food Engineering and Quality and Safety, Food (Edible Fungi) Processing Technology Research Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Jinchi Lan
- National Experimental Teaching Demonstration Center of Food Engineering and Quality and Safety, Food (Edible Fungi) Processing Technology Research Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yuxin Wang
- National Experimental Teaching Demonstration Center of Food Engineering and Quality and Safety, Food (Edible Fungi) Processing Technology Research Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Yuxiang Pan
- National Experimental Teaching Demonstration Center of Food Engineering and Quality and Safety, Food (Edible Fungi) Processing Technology Research Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Tianyuan Song
- National Experimental Teaching Demonstration Center of Food Engineering and Quality and Safety, Food (Edible Fungi) Processing Technology Research Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China
| | - Shizhu Liu
- Zhejiang Fangge Pharmaceutical Co. Ltd, Lishui, 323800, China
| | - Zhenyu Gu
- National Experimental Teaching Demonstration Center of Food Engineering and Quality and Safety, Food (Edible Fungi) Processing Technology Research Center, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, China.
| | - Yujun Ge
- Central blood station of Jiaxing, Jiaxing, 314000, China
| |
Collapse
|
6
|
Mustonen AM, Malinen M, Paakinaho V, Lehenkari P, Palosaari S, Kärjä V, Nieminen P. RNA sequencing analysis reveals distinct gene expression patterns in infrapatellar fat pads of patients with end-stage osteoarthritis or rheumatoid arthritis. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159576. [PMID: 39489461 DOI: 10.1016/j.bbalip.2024.159576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Osteoarthritis (OA) and autoimmune-driven rheumatoid arthritis (RA) are inflammatory joint diseases that share partly similar symptoms but have different, inadequately understood pathogeneses. Adipose tissues, including intra-articular infrapatellar fat pad (IFP), may contribute to their development. Analysis of differentially expressed genes (DEGs) in IFPs could improve the diagnostics of these conditions and help to develop novel treatment strategies. The aim was to identify potentially crucial genes and pathways discriminating OA and RA IFPs using RNA sequencing analysis. We aimed to distinguish genetically distinct patient groups as a starting point for further translational studies with the eventual goal of personalized medicine. Samples were collected from arthritic knees during total knee arthroplasty of sex- and age-matched OA and seropositive RA patients (n = 5-6/group). Metabolic pathways of interest were investigated by whole transcriptome sequencing, and DEGs were analyzed with univariate tests, hierarchical clustering (HC), and pathway analyses. There was significant interindividual variation in mRNA expression patterns, but distinct subgroups of OA and RA patients emerged that reacted similarly to their disease states based on HC. Compared to OA, RA samples showed 703 genes to be upregulated and 691 genes to be downregulated. Signaling pathway analyses indicated that these DEGs had common pathways in lipid metabolism, fatty acid biosynthesis and degradation, adipocytokine and insulin signaling, inflammatory response, and extracellular matrix organization. The divergent mRNA expression profiles in RA and OA suggest contribution of IFP to the regulation of synovial inflammatory processes and articular cartilage degradation and could provide novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; Department of Environmental and Biological Sciences, Faculty of Science, Forestry and Technology, University of Eastern Finland, P.O. Box 111, FI-80101 Joensuu, Finland.
| | - Marjo Malinen
- Department of Forestry and Environmental Engineering, South-Eastern Finland University of Applied Sciences, Paraatikenttä 7, FI-45100 Kouvola, Finland.
| | - Ville Paakinaho
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Petri Lehenkari
- Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland; Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland; Department of Surgery, Oulu University Hospital, P.O. Box 21, FI-90029 OYS, Finland.
| | - Sanna Palosaari
- Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland; Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland.
| | - Vesa Kärjä
- Department of Clinical Pathology, Kuopio University Hospital, Puijonlaaksontie 2, FI-70210 Kuopio, Finland.
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
7
|
Diao B, Fan Z, Zhou B, Zhan H. Crosstalk between pancreatic cancer and adipose tissue: Molecular mechanisms and therapeutic implications. Biochem Biophys Res Commun 2024; 740:151012. [PMID: 39561650 DOI: 10.1016/j.bbrc.2024.151012] [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: 08/03/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
The incidence rate of pancreatic cancer, a fatal illness with a meager 5-year survival rate, has been on the rise in recent times. When individuals accumulate excessive amounts of adipose tissue, the adipose organ becomes dysfunctional due to alterations in the adipose tissue microenvironment associated with inflammation and metabolism. This phenomenon may potentially contribute to the aberrant accumulation of fat that initiates pancreatic carcinogenesis, thereby influencing the disease's progression, resistance to treatment, and metastasis. This review presents a summary of the impact of pancreatic steatosis, visceral fat, cancer-associated adipocytes and lipid diets on the advancement of pancreatic cancer, as well as the reciprocal effects of pancreatic cancer on adipose tissue. Understanding the molecular mechanisms underlying the relationship between dysfunctional adipose tissue and pancreatic cancer better may lead to the discovery of new therapeutic targets for the disease's prevention and individualized treatment. This is especially important given the rising global incidence of obesity, which will improve the pancreatic cancer treatment options that are currently insufficient.
Collapse
Affiliation(s)
- Boyu Diao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
8
|
Li Y, Zeng Q, Peng D, Hu P, Luo J, Zheng K, Yin Y, Si R, Xiao J, Li S, Fu J, Liu J, Huang Y. Association of remnant cholesterol with insulin resistance and type 2 diabetes: mediation analyses from NHANES 1999-2020. Lipids Health Dis 2024; 23:404. [PMID: 39695677 DOI: 10.1186/s12944-024-02393-6] [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: 09/26/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Previous studies have established a correlation between elevated levels of remnant cholesterol (RC) and the occurrence of type 2 diabetes mellitus (T2D) as well as insulin resistance (IR); however, the precise nature of these associations remains incompletely elucidated. This study aimed to evaluate the relationships between RC and IR, as well as RC and T2D, and to determine the extent to which IR mediated the relationship between RC and T2D. METHODS This was an observational study that utilized cross-sectional methods to examine the general population in the National Health and Nutrition Examination Survey (NHANES) 1999-2020. The participants were divided into 4 groups according to the RC quartiles. The outcome was the prevalence of IR and T2D. Survey-weighted binary logistic regression analysis was used to analyze the associations, and the restricted cubic spline (RCS) curve was used to further analyze the nonlinear relationship. Receiver operating characteristic (ROC) curves were generated to evaluate the diagnostic performance, and the areas under the curves (AUC) of RC, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides (TG) were compared using the DeLong test. The mediating effect of IR on the relationship between RC and T2D was evaluated through mediation analysis. RESULTS A total of 23,755 participants (46.02 ± 18.48 years, 48.8% male) were included in our study. Higher RC levels were significantly associated with increased prevalence of both IR and T2D. After adjusting for potential confounders, logistic regression analysis showed that higher RC quartiles were associated with the increased prevalence of IR [Quartile 4 vs. Quartile 1: odds ratio (OR) (95% confidence interval, CI): 1.65 (1.41-1.94), p < 0.001] and T2D [Quartile 4 vs. Quartile 1: OR (95% CI): 1.24 (1.03-1.50), p = 0.024]. RCS analysis revealed two distinct nonlinear relationships: one between RC levels and the prevalence of IR (nonlinear p < 0.001), and another between RC levels and the prevalence of T2D (nonlinear p < 0.001). ROC curve analysis demonstrated that RC had the highest discriminative ability, significantly outperforming LDL-C, HDL-C, and TG in predicting both IR and T2D risk (all P < 0.001 by DeLong test). Mediation analysis revealed that IR significantly mediated the relationship between RC and T2D, with approximately 54.1% of the effect of RC on T2D being indirect through IR. CONCLUSIONS Higher RC level was associated with increased prevalence of IR and T2D. IR mediated 54.1% of the association between RC and T2D, suggesting that managing IR could be crucial in reducing the risk of T2D in individuals with elevated RC levels.
Collapse
Affiliation(s)
- Yuying Li
- School of Basic Medical Sciences, Capital medical university, Beijing, China
| | - Qiao Zeng
- School of Medical Technology and Nursing, Ji'an College, Ji'an, Jiangxi, China
| | - Danping Peng
- Department of Endocrinology, Ji'an Central Hospital, Ji'an, Jiangxi, China
| | - Pingsheng Hu
- Department of Respiratory and Critical Care Medicine, Ji'an Central Hospital, Ji'an, Jiangxi, China
| | - Jiahua Luo
- Department of Neurology, Ji'an Central Hospital, Ji'an, Jiangxi, China
| | - Keyang Zheng
- Department of General Practice, Beijing Nuclear Industry Hospital, Beijing, China
| | - Yuzhe Yin
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Rite Si
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, National Clinical Research Center for Mental Disorders &National Center for Mental Disorders, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jingyi Xiao
- The Sixth Clinical Medical School, Capital Medical University, Beijing, China
| | - Shaofen Li
- Department of Laboratory, Ji'an Central Hospital, Ji'an, Jiangxi, China
| | - Jinxiang Fu
- Department of Endocrinology, Ji'an Central Hospital, Ji'an, Jiangxi, China
| | - Jinping Liu
- Department of Endocrinology, Ji'an Central Hospital, Ji'an, Jiangxi, China
| | - Yuqing Huang
- Department of Endocrinology, Affiliated Hospital of Jinggangshan University, No.1,Quanshuiyan Road,Jizhou District, Ji'an City, 343000, Jiangxi Province, China.
| |
Collapse
|
9
|
Lee EO, Jin H, Kim S, Joo HK, Lee YR, An SY, Piao S, Lee KH, Jeon BH. Alterations in Adipose Tissue and Adipokines in Heterozygous APE1/Ref-1 Deficient Mice. Endocrinol Metab (Seoul) 2024; 39:932-945. [PMID: 39566547 PMCID: PMC11695485 DOI: 10.3803/enm.2024.2061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGRUOUND The role of apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) in adipose tissue remains poorly understood. This study investigates adipose tissue dysfunction in heterozygous APE1/Ref-1 deficiency (APE1/Ref-1+/-) mice, focusing on changes in adipocyte physiology, oxidative stress, adipokine regulation, and adipose tissue distribution. METHODS APE1/Ref-1 mRNA and protein levels in white adipose tissue (WAT) were measured in APE1/Ref-1+/- mice, compared to their wild-type (APE1/Ref-1+/+) controls. Oxidative stress was assessed by evaluating reactive oxygen species (ROS) levels. Histological and immunohistochemical analyses were conducted to observe adipocyte size and macrophage infiltration of WAT. Adipokine expression was measured, and micro-magnetic resonance imaging (MRI) was used to quantify abdominal fat volumes. RESULTS APE1/Ref-1+/- mice exhibited significant reductions in APE1/Ref-1 mRNA and protein levels in WAT and liver tissue. These mice also showed elevated ROS levels, suggesting a regulatory role for APE1/Ref-1 in oxidative stress in WAT and liver. Histological and immunohistochemical analyses revealed hypertrophic adipocytes and macrophage infiltration in WAT, while Oil Red O staining demonstrated enhanced ectopic fat deposition in the liver of APE1/Ref-1+/- mice. These mice also displayed altered adipokine expression, with decreased adiponectin and increased leptin levels in the WAT, along with corresponding alterations in plasma levels. Despite no significant changes in overall body weight, microMRI assessments demonstrated a significant increase in visceral and subcutaneous abdominal fat volumes in APE1/Ref-1+/- mice. CONCLUSION APE1/Ref-1 is crucial in adipokine regulation and mitigating oxidative stress. These findings suggest its involvement in adipose tissue dysfunction, highlighting its potential impact on abdominal fat distribution and its implications for obesity and oxidative stress-related conditions.
Collapse
Affiliation(s)
- Eun-Ok Lee
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hao Jin
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Sungmin Kim
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hee Kyoung Joo
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yu Ran Lee
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Soo Yeon An
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Division of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shuyu Piao
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kwon Ho Lee
- Department of Physical Therapy, Joongbu University, Geumsan, Korea
| | - Byeong Hwa Jeon
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
10
|
Danowska M, Stefanowicz M, Strączkowski M. The expression of NFAT family genes in subcutaneous adipose tissue before and after weight loss in obese individuals. Nutr Metab Cardiovasc Dis 2024; 34:2455-2463. [PMID: 39069466 DOI: 10.1016/j.numecd.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND AIMS Adipose tissue (AT) serves as a vital energy storage site and plays a pivotal role in metabolic regulation, exhibiting a high response to insulin. Impairment in this response may closely associate with obesity, and NFAT (nuclear factor of activated T cells) family genes may be involved in the process. However, human data linking NFAT and AT remains elusive. The aim of this study was to assess the expression of NFAT family genes and markers of adipogenesis in subcutaneous adipose tissue (SAT) among normal-weight and overweight/obese individuals before and after weight loss, in relation to insulin sensitivity. METHODS AND RESULTS The study included 45 participants, 15 normal-weight (control group) and 30 overweight or obese, who underwent a 12-week dietary intervention (DI) program. Before and after the program hyperinsulinemic-euglycemic clamp and SAT biopsy were conducted. Before DI, a positive correlations was observed in the expression of NFATc1, NFATc4, and NFAT5 with insulin sensitivity. The expression of NFAT family genes and markers of adipogenesis in SAT was lower in individuals with overweight or obesity compared to normal-weight. Additionally, a positive correlation was noted between NFAT family genes and adipogenesis markers both before and after weight loss. Following the DI program, there was an increase in the expression of NFATc3, NFATc4, and NFAT5 in SAT. CONCLUSION Decreased SAT expression of NFAT genes in obesity is partly reversed in response to weight loss. NFAT genes in SAT are associated with insulin sensitivity and adipogenesis. Registration number for clinical trial: NCT01393210.
Collapse
Affiliation(s)
- Magdalena Danowska
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Magdalena Stefanowicz
- Department of Metabolic Diseases, Medical University of Bialystok, Bialystok, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland.
| |
Collapse
|
11
|
Qu J, Tian L, Zhang M, Sun B, Chen L. SGLT2 inhibitor canagliflozin reduces visceral adipose tissue in db/db mice by modulating AMPK/KLF4 signaling and regulating mitochondrial dynamics to induce browning. Mol Cell Endocrinol 2024; 592:112320. [PMID: 38964727 DOI: 10.1016/j.mce.2024.112320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Obesity is characterized by excessive accumulation of adipose tissue (mainly visceral). The morphology and function of mitochondria are crucial for regulating adipose browning and weight loss. Research suggests that the SGLT2 inhibitor canagliflozin may induce weight loss through an unknown mechanism, particularly targeting visceral adipose tissue. While Krueppel-Like Factor 4 (KLF4) is known to be essential for energy metabolism and mitochondrial function, its specific impact on visceral adipose tissue remains unclear. We administered canagliflozin to db/db mice for 8 weeks, or exposed adipocytes to canagliflozin for 24 h. The expression levels of browning markers, mitochondrial dynamics, and KLF4 were assessed. Then we validated the function of KLF4 through overexpression in vivo and in vitro. Adenosine monophosphate-activated protein kinase (AMPK) agonists, inhibitors, and KLF4 si-RNA were employed to elucidate the relationship between AMPK and KLF4. The findings demonstrated that canagliflozin significantly decreased body weight in db/db mice and augmented cold-induced thermogenesis. Additionally, canagliflozin increased the expression of mitochondrial fusion-related factors while reducing the levels of fission markers in epididymal white adipose tissue. These consistent findings were mirrored in canagliflozin-treated adipocytes. Similarly, overexpression of KLF4 in both adipocytes and db/db mice yielded comparable results. In all, canagliflozin mitigates obesity in db/db mice by promoting the brown visceral adipocyte phenotype through enhanced mitochondrial fusion via AMPK/KLF4 signaling.
Collapse
Affiliation(s)
- Jingru Qu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Lei Tian
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Man Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China.
| |
Collapse
|
12
|
Shen ZJ, Liu M, Zhang JX, Huang Y, Kong Y, Liu SG. Comparison of Serum Vitamin D Levels in Obese Subjects with and without Type 2 Diabetes Mellitus. J Inflamm Res 2024; 17:5915-5922. [PMID: 39247834 PMCID: PMC11378779 DOI: 10.2147/jir.s475180] [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: 04/23/2024] [Accepted: 07/20/2024] [Indexed: 09/10/2024] Open
Abstract
Background Although observational studies have reported a correlation between vitamin D deficiency and type 2 diabetes mellitus (T2DM), epidemiological evidence on the risk of obese subjects suffering T2DM due to a vitamin D deficiency is limited. Therefore, we investigated the correlation between T2DM and serum vitamin D, lipids, blood pressure, insulin indexes in an obese population. Methods A total of 1440 participants including 450 healthy controls and 990 obese subjects, 470 without T2DM and 520 with T2DM. Serum vitamin D levels were measured, and the association between low levels and T2DM in obese subjects was examined using multinomial and linear regression analyses. Results Of the participants, 35% had deficient or insufficient vitamin D levels (ie, <20 ng/mL). Compared with healthy controls, obese subjects, particularly those with T2DM had lower vitamin D levels. Multinomial logistic regression analysis showed that obese subjects with T2DM had a gradually increasing risk for desirable (RO = 1.41, 95% CI 1.06-1.93, P = 0.027), insufficient (RO = 1.83, 95% CI 1.27-2.84, P < 0.001), or deficient ((RO = 2.14, 95% CI 1.15-3.75, P = 0.014) vitamin D levels. In obese subjects with T2DM, vitamin D levels correlated inversely with the risk indicators for diabetes, such as the levels of HbA1c (β = -0.16, P = 0.002), fasting insulin (Fins; β = -0.31, P = 0.008), and HOMA-IR (β = -0.19, P < 0.001). In obese subjects without T2DM, vitamin D was associated negatively with the risk of having T2DM at five-year follow-up (relative risk = 0.93, 95% CI 0.79-0.97, P = 0.037). Conclusion This study demonstrates that low vitamin D levels correlate with the presence of T2DM in the obese population. This finding indicates that hypovitaminosis D may be a potential biological vulnerability factor for the development of T2DM in obese subjects.
Collapse
Affiliation(s)
- Zhi-Jun Shen
- Department of Clinical Laboratory, Hubei Third People´s Hospital Affiliated to Jianghan University, Wuhan, 430033, People's Republic of China
| | - Miao Liu
- Department of Clinical Laboratory, Hubei Third People´s Hospital Affiliated to Jianghan University, Wuhan, 430033, People's Republic of China
| | - Jun-Xia Zhang
- Department of Clinical Laboratory, Hubei Third People´s Hospital Affiliated to Jianghan University, Wuhan, 430033, People's Republic of China
| | - Yu Huang
- Department of Clinical Laboratory, Hubei Third People´s Hospital Affiliated to Jianghan University, Wuhan, 430033, People's Republic of China
| | - Ying Kong
- Department of Clinical Laboratory, Hubei Third People´s Hospital Affiliated to Jianghan University, Wuhan, 430033, People's Republic of China
| | - Shi-Guo Liu
- Department of Clinical Laboratory, Hubei Third People´s Hospital Affiliated to Jianghan University, Wuhan, 430033, People's Republic of China
| |
Collapse
|
13
|
Batitucci G, Almeida OG, De Martinis ECP, Solar I, Cintra DE, de Freitas EC. Intermittent fasting and high-intensity interval training do not alter gut microbiota composition in adult women with obesity. Am J Physiol Endocrinol Metab 2024; 327:E241-E257. [PMID: 38922577 DOI: 10.1152/ajpendo.00310.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024]
Abstract
Obesity is advancing at an accelerated pace, and yet its treatment is still an emerging field. Although studies have demonstrated the role of the microbiota in the pathogenesis of obesity, this is the first study to show the effects of intermittent fasting (IF), combined or not with exercise, and high-intensity interval training (HIIT) on the gut microbiota composition in women with obesity. Our hypothesis is that IF combined with HIIT can promote the remodeling of the composition and function of the gut microbiota. Thirty-six women with obesity, aged between 18 and 40 yr, participated in the study. They were randomly divided into three groups: 1) IF associated with HIIT group [IF + exercise group (EX), n = 15]; 2) HIIT group (EX, n = 11); and 3) IF group (IF, n = 10). Interventions took place over 8 wk, and all assessments were performed preintervention and postintervention. The HIIT circuit was performed 3 times/wk, for 25 min/session. The IF protocol was a 5:2 (2 times/wk). Multiplex analysis of inflammatory cytokines, sequencing of the 16S rRNA gene, and gas chromatography to measure fecal concentrations of short-chain fatty acids (SCFAs) were performed. This study was registered on ClinicalTrials.gov (NCT05237154). Exercise increased fecal acetate concentrations (P = 0.04), but no changes were observed in the composition and functional profile of the microbiota. The interventions did not change the composition of the microbiota, but exercise may play a modulatory role in the production of acetate. This investigation provides clinical insights into the use of IF and HIIT for women with obesity.NEW & NOTEWORTHY This is the first investigation about alternate-day fasting combined with HITT on the gut microbiota of obese women. The study contributes to the advancement of human science involving IF and HIIT, popular strategies for managing obesity. Previous evidence has explored IF in modulating the microbiota in animal models or specific populations and clinical conditions. Despite the subtle outcomes, this study has relevance and originality in the field of gut microbiota knowledge.
Collapse
Affiliation(s)
- Gabriela Batitucci
- Department of Food and Nutrition, School of Pharmaceutical Sciences of Araraquara, State University of Sao Paulo, Araraquara, Brazil
| | - Otávio G Almeida
- Ribeirão Preto School of Pharmaceutical Sciences, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Elaine C P De Martinis
- Ribeirão Preto School of Pharmaceutical Sciences, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Isabela Solar
- Faculty of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Dennys E Cintra
- Nutritional Genomics Laboratory and Nutrigenomics and Lipids Center, Faculty of Applied Sciences, University of Campinas, Limeira, Brazil
| | - Ellen Cristini de Freitas
- Department of Food and Nutrition, School of Pharmaceutical Sciences of Araraquara, State University of Sao Paulo, Araraquara, Brazil
- Laboratory of Exercise Physiology and Metabolism, School of Physical Education and Sports of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
14
|
Esteves JV, Stanford KI. Exercise as a tool to mitigate metabolic disease. Am J Physiol Cell Physiol 2024; 327:C587-C598. [PMID: 38981607 PMCID: PMC11427015 DOI: 10.1152/ajpcell.00144.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Metabolic diseases, notably obesity and type 2 diabetes (T2D), have reached alarming proportions and constitute a significant global health challenge, emphasizing the urgent need for effective preventive and therapeutic strategies. In contrast, exercise training emerges as a potent intervention, exerting numerous positive effects on metabolic health through adaptations to the metabolic tissues. Here, we reviewed the major features of our current understanding with respect to the intricate interplay between metabolic diseases and key metabolic tissues, including adipose tissue, skeletal muscle, and liver, describing some of the main underlying mechanisms driving pathogenesis, as well as the role of exercise to combat and treat obesity and metabolic disease.
Collapse
Affiliation(s)
- Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
15
|
Saglam E, Karagedik H, Dinc M, Oke D, Gun Atak P, Karadeniz B, Burul G, Gormus Degrigo U. Can Bone Morphogenetic Protein 1 (BMP1) Be a Potential Biomarker of Obesity? Cureus 2024; 16:e67025. [PMID: 39280566 PMCID: PMC11402472 DOI: 10.7759/cureus.67025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 09/18/2024] Open
Abstract
Background Obesity has long been a severe threat to public health as an epidemic, and studies on its pathogenesis and treatment have been ongoing. Our study aims to compare the serum levels of bone morphogenetic protein 1 (BMP1), neuregulin 4 (NRG4), and apolipoprotein A5 (ApoA5) in obese and non-obese individuals and investigate their association with obesity. Methodology Our study included a total of 111 participants, of whom 46 were obese (body mass index (BMI) ≥30 kg/m2), aged 18-65 years, and had no comorbidities, and 65 were non-obese (BMI = 18.5-29.9 kg/m2) without any additional disease. For all participants, BMP1, NRG4, and ApoA5 levels were determined and compared with clinical and biochemical parameters. Results Overall, 60.4% (n = 67) of the participants were female and 39.6% (n = 44) were male. In terms of the BMI scores, 58.6% (n = 65) had a BMI <30 kg/m2 and 41.4% (n = 46) had a BMI ≥30 kg/m2. Both, the BMI and the gender groups did not differ significantly in terms of age (p = 0.093 and p = 0.795, respectively). The weight, fat-free mass, mineral quantity, protein quantity, fluid weight, and fluid ratio values of the male participants were significantly higher than females (p = 0.011, p = 0.001, p = 0.001, p = 0.001, p = 0.001, and p = 0.001, respectively). The aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratios and the triglyceride/glucose (TG/Glu) ratios were found to be significantly higher in males than in females (p = 0.001 and p = 0.001, respectively). The respective BMP1 (15.88 vs. 13.35), AST/ALT (1.36 vs. 1.04) and TG/Glu ratios (1.47 vs. 1.29) were significantly higher, while the quantitative insulin sensitivity check index (QUICKI) was lower in obese individuals than in non-obese individuals (0.32 vs. 0.34). NRG4 and ApoA5 values were similar between the two groups. BMP1, QUICKI values, and AST/ALT ratios proved to be statistically significant in obesity through the univariable logistic regression analysis (β = 1.066, p = 0.048; β = 0.0001, p = 0.001, and β = 3.707, p = 0.003, respectively). On multiple logistic regression analysis, QUICKI values (β = 0.001, p = 0.001) had a negative and significant effect on obesity, and the AST/ALT ratios (β = 2.803, p = 0.033) had a positive and significant effect on obesity. Conclusions Our study indicates that detecting an important link between BMP1 in obese patients will help elucidate the pathogenesis of obesity and come up with a potential therapeutic candidate. BMP1 levels, along with AST/ALT and TG/Glu ratios, were significantly higher in obese patients. BMP1 levels were also an independent significant predictor of obesity together with AST/ALT ratio and QUICKI in this study, suggesting that it may exhibit a metabolic deterioration in obese individuals. However, the results cannot absolutely tell whether it supported deterioration or was a component of the repair mechanism. Althoughit is generally known from recent studies that BMP1 plays a role in osteogenesis, some encouraging results were obtained in our study indicating that BMP1 may play a role in the pathogenesis of obesity. It is expected that our results will not only promote the elucidation of the pathogenesis of obesity, but also provide a therapeutic agent.
Collapse
Affiliation(s)
- Emel Saglam
- Internal Medicine, Bagcilar Training and Research Hospital, Istanbul, TUR
| | - Hande Karagedik
- Molecular Medicine, Aziz Sancar Institute for Experimental Medicine, Istanbul, TUR
| | - Mustafa Dinc
- Endocrinology and Metabolism, Kirklareli Training and Research Hospital, Kirklareli, TUR
| | - Deniz Oke
- Physical Medicine and Rehabilitation, Gaziosmanpasa Training and Research Hospital, Istanbul, TUR
| | | | - Burcak Karadeniz
- Rheumatology, Bagcilar Training and Research Hospital, Istanbul, TUR
| | - Gokhan Burul
- Internal Medicine, Bagcilar Training and Research Hospital, Istanbul, TUR
| | | |
Collapse
|
16
|
Wu S, Teng Y, Lan Y, Wang M, Zhang T, Wang D, Qi F. The association between fat distribution and α1-acid glycoprotein levels among adult females in the United States. Lipids Health Dis 2024; 23:235. [PMID: 39080765 PMCID: PMC11290176 DOI: 10.1186/s12944-024-02223-9] [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: 03/26/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Visceral fat accumulation and obesity-induced chronic inflammation have been proposed as early markers for multiple disease states, especially in women. Nevertheless, the potential impact of fat distribution on α1-acid glycoprotein(AGP), a marker of inflammation, remains unclear. This research was conducted to investigate the relationships among obesity, fat distribution, and AGP levels. METHODS A cross-sectional observational study was performed using blood samples from adult females recruited through the National Health and Nutrition Examination Survey from 2015 to 2018. Serum levels of AGP were measured using the Tina-quant α-1-Acid Glycoprotein Gen.2 assay. Based on the fat distribution data obtained from dual-energy X-ray absorptiometry assessments, body mass index (BMI), total percent fat (TPF), android percent fat (APF), gynoid percent fat (GPF), android fat/gynoid fat ratio (AGR), visceral percent fat (VPF), subcutaneous percent fat (SPF), visceral fat/subcutaneous fat ratio (VSR) were used as dependent variables. To investigate the link between fat distribution and AGP, multivariate linear regression analysis was utilized. Furthermore, a sensitivity analysis was also performed. RESULTS The present study included 2,295 participants. After adjusting for covariates, BMI, TPF, APF, GPF, VPF, and SPF were found to be positively correlated with AGP levels (BMI: β = 23.65 95%CI:20.90-26.40; TPF: β = 25.91 95%CI:23.02-28.80; APF: β = 25.21 95%CI:22.49-27.93; GPF: β = 19.65 95%CI:16.96-22.34; VPF: β = 12.49 95%CI:9.08-15.90; SPF: β = 5.69, 95%CI:2.89-8.49; AGR: β = 21.14 95%CI:18.16-24.12; VSR: β = 9.35 95%CI:6.11-12.59, all P < 0.0001). All the above indicators exhibited a positive dose-response relationship with AGP. In terms of fat distribution, both AGR and VSR showed positive associations with AGP (P for trend < 0.0001). In particular, when compared to individuals in tertile 1 of AGR, participants in tertiles 2 and 3 had 13.42 mg/dL (95% CI 10.66-16.18) and 21.14 mg/dL (95% CI 18.16-24.12) higher AGP levels, respectively. Participants in the highest tertile of VSR were more likely to exhibit a 9.35 mg/dL increase in AGP compared to those in the lowest tertile (95% CI 6.11-12.59). CONCLUSIONS Overall, this study revealed a positive dose-dependent relationship between fat proportion/distribution and AGP levels in women. These findings suggest that physicians can associate abnormal serum AGP and obesity with allow timely interventions.
Collapse
Affiliation(s)
- Siqi Wu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Ying Teng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Yuanqi Lan
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Maoyang Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Tianhua Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China
| | - Dali Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| | - Fang Qi
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563000, People's Republic of China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
17
|
Minato-Inokawa S, Honda M, Tsuboi-Kaji A, Takeuchi M, Kitaoka K, Kurata M, Wu B, Kazumi T, Fukuo K. Adipose tissue insulin resistance index was inversely associated with gluteofemoral fat and skeletal muscle mass in Japanese women. Sci Rep 2024; 14:16347. [PMID: 39013950 PMCID: PMC11252386 DOI: 10.1038/s41598-024-67184-6] [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: 04/28/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Associations of adipose tissue insulin resistance index (AT-IR, a product of fasting insulin and free fatty acids) with body fat mass and distribution and appendicular skeletal muscle mass (ASM) were compared with results of homeostasis-model assessment-insulin resistance (HOMA-IR) in 284 Japanese female university students and 148 their biological mothers whose BMI averaged < 23 kg/m2. Although mothers compared with daughters had higher BMI, body fat percentage, trunk fat to body fat (TF/BF) ratio and lower leg fat to body fat (LF/BF), AT-IR and HOMA-IR did not differ. We had multivariable linear regression analyses which included TF/BF ratio, LF/BF ratio, weight-adjusted ASM (%ASM), height-adjusted ASM index (ASMI), fat mass index (FMI), and body fat percentage. In young women, AT-IR was independently associated with LF/BF ratio (Standardized β [Sβ]: - 0.139, p = 0.019) and ASMI (Sβ: - 0.167, p = 0.005). In middle-aged women, LF/BF ratio (Sβ: - 0.177, p = 0.049) and %ASM (Sβ: - 0.205, p = 0.02) emerged as independent determinants of AT-IR. HOMA-IR was associated with TF/BF ratio and FMI, a proxy of abdominal and general adiposity, respectively, in both young and middle-aged women. The inverse association of AT-IR with leg fat may support the notion that limited peripheral adipose storage capacity and small skeletal muscle size are important etiological components in insulin-resistant cardiometabolic disease in Japanese women.
Collapse
Affiliation(s)
- Satomi Minato-Inokawa
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Laboratory of Community Health and Nutrition, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Ehime, Japan
| | - Mari Honda
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, Hyogo, Japan
| | - Ayaka Tsuboi-Kaji
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Nutrition, Osaka City Juso Hospital, Osaka, Japan
| | - Mika Takeuchi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
| | - Kaori Kitaoka
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Advanced Epidemiology, Noncommunicable Disease (NCD) Epidemiology Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Miki Kurata
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Bin Wu
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Tsutomu Kazumi
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan.
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan.
- Department of Medicine, Kohan Kakogawa Hospital, Kakogawa, Hyogo, Japan.
| | - Keisuke Fukuo
- Research Institute for Nutrition Sciences, Mukogawa Women's University, 6-46, Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
- Open Research Center for Studying of Lifestyle-Related Diseases, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
- Department of Food Sciences and Nutrition, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
18
|
Talamonti E, Davegardh J, Kalinovich A, van Beek SMM, Dehvari N, Halleskog C, Bokhari HM, Hutchinson DS, Ham S, Humphrys LJ, Dijon NC, Motso A, Sandstrom A, Zacharewicz E, Mutule I, Suna E, Spura J, Ditrychova K, Stoddart LA, Holliday ND, Wright SC, Lauschke VM, Nielsen S, Scheele C, Cheesman E, Hoeks J, Molenaar P, Summers RJ, Pelcman B, Yakala GK, Bengtsson T. The novel adrenergic agonist ATR-127 targets skeletal muscle and brown adipose tissue to tackle diabesity and steatohepatitis. Mol Metab 2024; 85:101931. [PMID: 38796310 PMCID: PMC11258667 DOI: 10.1016/j.molmet.2024.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/09/2024] [Accepted: 03/29/2024] [Indexed: 05/28/2024] Open
Abstract
OBJECTIVE Simultaneous activation of β2- and β3-adrenoceptors (ARs) improves whole-body metabolism via beneficial effects in skeletal muscle and brown adipose tissue (BAT). Nevertheless, high-efficacy agonists simultaneously targeting these receptors whilst limiting activation of β1-ARs - and thus inducing cardiovascular complications - are currently non-existent. Therefore, we here developed and evaluated the therapeutic potential of a novel β2-and β3-AR, named ATR-127, for the treatment of obesity and its associated metabolic perturbations in preclinical models. METHODS In the developmental phase, we assessed the impact of ATR-127's on cAMP accumulation in relation to the non-selective β-AR agonist isoprenaline across various rodent β-AR subtypes, including neonatal rat cardiomyocytes. Following these experiments, L6 muscle cells were stimulated with ATR-127 to assess the impact on GLUT4-mediated glucose uptake and intramyocellular cAMP accumulation. Additionally, in vitro, and in vivo assessments are conducted to measure ATR-127's effects on BAT glucose uptake and thermogenesis. Finally, diet-induced obese mice were treated with 5 mg/kg ATR-127 for 21 days to investigate the effects on glucose homeostasis, body weight, fat mass, skeletal muscle glucose uptake, BAT thermogenesis and hepatic steatosis. RESULTS Exposure of L6 muscle cells to ATR-127 robustly enhanced GLUT4-mediated glucose uptake despite low intramyocellular cAMP accumulation. Similarly, ATR-127 markedly increased BAT glucose uptake and thermogenesis both in vitro and in vivo. Prolonged treatment of diet-induced obese mice with ATR-127 dramatically improved glucose homeostasis, an effect accompanied by decreases in body weight and fat mass. These effects were paralleled by an enhanced skeletal muscle glucose uptake, BAT thermogenesis, and improvements in hepatic steatosis. CONCLUSIONS Our results demonstrate that ATR-127 is a highly effective, novel β2- and β3-ARs agonist holding great therapeutic promise for the treatment of obesity and its comorbidities, whilst potentially limiting cardiovascular complications. As such, the therapeutic effects of ATR-127 should be investigated in more detail in clinical studies.
Collapse
Affiliation(s)
| | - Jelena Davegardh
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | | | - Nodi Dehvari
- Atrogi AB, Tomtebodavagen 6, Solna, Stockholm, Sweden
| | | | | | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Seungmin Ham
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Laura J Humphrys
- School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Nicola C Dijon
- School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Aikaterini Motso
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | | | - Evelyn Zacharewicz
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ilga Mutule
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Edgars Suna
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Jana Spura
- Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Karolina Ditrychova
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Leigh A Stoddart
- Excellerate Bioscience, The Triangle, NG2 Business Park, Nottingham, UK
| | - Nicholas D Holliday
- School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK; Excellerate Bioscience, The Triangle, NG2 Business Park, Nottingham, UK
| | - Shane C Wright
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Volker M Lauschke
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden; Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; Tübingen University, Tübingen, Germany
| | - Soren Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Righospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Elizabeth Cheesman
- Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Peter Molenaar
- Cardio-Vascular Molecular & Therapeutics Translational Research Group, Northside Clinical School of Medicine, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Queensland University of Technology (QUT), School of Biomedical Sciences, Institute of Health and Biomedical Innovation, 60 Musk Avenue, Kelvin Grove, Queensland, Australia
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | | | | | - Tore Bengtsson
- Atrogi AB, Tomtebodavagen 6, Solna, Stockholm, Sweden; Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
19
|
Chen Z, Qiu X, Wang Q, Wu J, Li M, Niu W. Serum vitamin D and obesity among US adolescents, NHANES 2011-2018. Front Pediatr 2024; 12:1334139. [PMID: 38836246 PMCID: PMC11148364 DOI: 10.3389/fped.2024.1334139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/29/2024] [Indexed: 06/06/2024] Open
Abstract
Background and objectives Childhood obesity is highly prevalent worldwide. We aimed to assess whether serum 25-hydroxyvitamin D was associated with general/central obesity among US adolescents, and further to explore the mediatory impact of homeostasis model assessment of insulin resistance (HOMA-IR) on this association. Methods This study is cross-sectional in design. Study adolescents were enrolled from the National Health and Nutrition Examination Survey (NHANES), 2011-2018. Serum 25-hydroxyvitamin D categories associated with general (indexed by body mass index) and central (indexed by waist circumference to height ratio) obesity were regressed. The possible mediatory effect of HOMA-IR on this association was explored. The nonlinear and dose-response association was examined by restricted cubic spline (RCS) test. Results Total 2,696 adolescents were eligible for inclusion, and the mean age of all adolescents was 15.4 years. Overall, the percentage of general and central obesity was 38.0% and 38.6%, respectively. Compared with adolescents with sufficient vitamin D, adolescent with deficient and insufficient vitamin D intake were associated with general obesity and central obesity; fully-adjusted OR for general obesity was 1.602 (95% CI: 1.161-2.211) and 1.659 (1.385-1.986), and fully-adjusted OR for central obesity was 2.025 (1.445-2.837) and 1.557 (1.287-1.884), respectively, while there was no observable significance in adolescents with possibly harmful vitamin D. The proportion mediated by HOMA-IR was estimated to be 31.7% for global obesity and 50.3% for central obesity (both P < 0.05). More stratified analyses were presented, and identified that the association with general obesity was particularly present among Mexican American, while with central obesity among Non-Hispanic Black adolescents. Conclusions Our findings indicate that deficient or insufficient 25-hydroxyvitamin D concentrations were associated with the significant risk of general and central obesity among US adolescents, and approximately 30% and 50%, respectively, of these associations were mediated by HOMA-IR.
Collapse
Affiliation(s)
- Zisu Chen
- Department of Pediatrics, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaojin Qiu
- Department of Pediatrics, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Qiong Wang
- Department of Pediatrics, China-Japan Friendship Hospital, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Wu
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing, China
| | - Min Li
- Department of Pediatrics, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wenquan Niu
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
20
|
Deehan EC, Mocanu V, Madsen KL. Effects of dietary fibre on metabolic health and obesity. Nat Rev Gastroenterol Hepatol 2024; 21:301-318. [PMID: 38326443 DOI: 10.1038/s41575-023-00891-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 02/09/2024]
Abstract
Obesity and metabolic syndrome represent a growing epidemic worldwide. Body weight is regulated through complex interactions between hormonal, neural and metabolic pathways and is influenced by numerous environmental factors. Imbalances between energy intake and expenditure can occur due to several factors, including alterations in eating behaviours, abnormal satiation and satiety, and low energy expenditure. The gut microbiota profoundly affects all aspects of energy homeostasis through diverse mechanisms involving effects on mucosal and systemic immune, hormonal and neural systems. The benefits of dietary fibre on metabolism and obesity have been demonstrated through mechanistic studies and clinical trials, but many questions remain as to how different fibres are best utilized in managing obesity. In this Review, we discuss the physiochemical properties of different fibres, current findings on how fibre and the gut microbiota interact to regulate body weight homeostasis, and knowledge gaps related to using dietary fibres as a complementary strategy. Precision medicine approaches that utilize baseline microbiota and clinical characteristics to predict individual responses to fibre supplementation represent a new paradigm with great potential to enhance weight management efficacy, but many challenges remain before these approaches can be fully implemented.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Nebraska Food for Health Center, Lincoln, NE, USA
| | - Valentin Mocanu
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
21
|
Flori L, Piragine E, Calderone V, Testai L. Role of hydrogen sulfide in the regulation of lipid metabolism: Implications on cardiovascular health. Life Sci 2024; 341:122491. [PMID: 38336275 DOI: 10.1016/j.lfs.2024.122491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/29/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The World Health Organization (WHO) defines obesity as an urgency for health and a social emergency. Today around 39 % of people is overweight, of these over 13 % is obese. It is well-consolidated that the adipose cells are deputy to lipid storage under caloric excess; however, despite the classical idea that adipose tissue has exclusively a passive function, now it is known to be deeply involved in the regulation of systemic metabolism in physiological as well as under obesogenic conditions, with consequences on cardiovascular health. Beside two traditional types of adipose cells (white and brown), recently the beige one has been highlighted as the consequence of the healthy remodeling of white adipocytes, confirming their metabolic adaptability. In this direction, pharmacological, nutraceutical and nutrient-based approaches are addressed to positively influence inflammation and metabolism, thus contributing to reduce the obese-associated cardiovascular risk. In this scenario, hydrogen sulfide emerges as a new mediator that may regulate crucial targets involved in the regulation of metabolism. The current evidence demonstrates that hydrogen sulfide may induce peroxisome proliferator activated receptor γ (PPARγ), a crucial mediator of adipogenesis, inhibit the phosphorylation of perlipin-1 (plin-1), a protein implicated in the lipolysis, and finally promote browning process, through the release of irisin from skeletal muscle. The results summarized in this review suggest an important role of hydrogen sulfide in the regulation of metabolism and in the prevention/treatment of obese-associated cardiovascular diseases and propose new insight on the putative mechanisms underlying the release of hydrogen sulfide or its biosynthesis, delineating a further exciting field of application.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Eugenia Piragine
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| | - Lara Testai
- Department of Pharmacy, University of Pisa, via Bonanno, 6-56120 Pisa, Italy.
| |
Collapse
|
22
|
Nishijima T, Yamashita Y, Ashida H. Black soybean seed coat polyphenols have different effects on glucose and lipid metabolism in growing and young adult mice. Food Funct 2024; 15:1004-1020. [PMID: 38180075 DOI: 10.1039/d3fo04269c] [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: 01/06/2024]
Abstract
Black soybean contains flavan-3-ols and cyanidin 3-O-glucoside in its seed coat. Polyphenol-rich black soybean seed coat extract (BE) possesses various health benefits, such as antioxidant, anti-obesity, and anti-hyperglycemic effects. However, these functions have been evaluated mainly in the growing stage of animals, and there is no comparison data for different life stages. In this present study, we compared the effect of BE in growing (5-week old) and young adult (22-week old) ICR male mice. These mice were given an AIN 93M diet containing 2.0% BE for 4 weeks. BE did not affect body weight gain in both growing and young adult mice, but it suppressed mesenteric and subcutaneous white adipose tissue weights and decreased the cell size. BE also significantly suppressed plasma free-fatty acid levels. The effect of both BE and life stages were observed in the protein expression of adipogenesis-related transcription factors; in particular, BE suppressed the expression of C/EBPα and PPARγ. No significant change was observed in lipolysis and lipogenesis factors in the white adipose tissue and liver. Alternatively, BE showed low glucose tolerance without affecting plasma insulin levels after glucose loading in young adult mice, as seen from the results of the oral glucose tolerance test. However, plasma glucose and insulin levels remained unchanged at the end of the experimental period. In conclusion, these results strongly suggest that the health-beneficial effects of BE may alter in mice at different life stages.
Collapse
Affiliation(s)
- Toshiki Nishijima
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-ku, Kobe, Hyogo 657-8501, Japan.
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-ku, Kobe, Hyogo 657-8501, Japan.
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-ku, Kobe, Hyogo 657-8501, Japan.
| |
Collapse
|
23
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
24
|
Kueck PJ, Morris JK, Stanford JA. Current Perspectives: Obesity and Neurodegeneration - Links and Risks. Degener Neurol Neuromuscul Dis 2023; 13:111-129. [PMID: 38196559 PMCID: PMC10774290 DOI: 10.2147/dnnd.s388579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
Obesity is increasing in prevalence across all age groups. Long-term obesity can lead to the development of metabolic and cardiovascular diseases through its effects on adipose, skeletal muscle, and liver tissue. Pathological mechanisms associated with obesity include immune response and inflammation as well as oxidative stress and consequent endothelial and mitochondrial dysfunction. Recent evidence links obesity to diminished brain health and neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). Both AD and PD are associated with insulin resistance, an underlying syndrome of obesity. Despite these links, causative mechanism(s) resulting in neurodegenerative disease remain unclear. This review discusses relationships between obesity, AD, and PD, including clinical and preclinical findings. The review then briefly explores nonpharmacological directions for intervention.
Collapse
Affiliation(s)
- Paul J Kueck
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jill K Morris
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - John A Stanford
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| |
Collapse
|
25
|
Reed RM, Whyte MB, Goff LM. Cardiometabolic disease in Black African and Caribbean populations: an ethnic divergence in pathophysiology? Proc Nutr Soc 2023:1-11. [PMID: 38230432 DOI: 10.1017/s0029665123004895] [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: 01/18/2024]
Abstract
In the UK, populations of Black African and Caribbean (BAC) ethnicity suffer higher rates of cardiometabolic disease than White Europeans (WE). Obesity, leading to increased visceral adipose tissue (VAT) and intrahepatic lipid (IHL), has long been associated with cardiometabolic risk, driving insulin resistance and defective fatty acid/lipoprotein metabolism. These defects are compounded by a state of chronic low-grade inflammation, driven by dysfunctional adipose tissue. Emerging evidence has highlighted associations between central complement system components and adipose tissue, fatty acid metabolism and inflammation; it may therefore sit at the intersection of various cardiometabolic disease risk factors. However, increasing evidence suggests an ethnic divergence in pathophysiology, whereby current theories fail to explain the high rates of cardiometabolic disease in BAC populations. Lower fasting and postprandial TAG has been reported in BAC, alongside lower VAT and IHL deposition, which are paradoxical to the high rates of cardiometabolic disease exhibited by this ethnic group. Furthermore, BAC have been shown to exhibit a more anti-inflammatory profile, with lower TNF-α and greater IL-10. In contrast, recent evidence has revealed greater complement activation in BAC compared to WE, suggesting its dysregulation may play a greater role in the high rates of cardiometabolic disease experienced by this population. This review outlines the current theories of how obesity is proposed to drive cardiometabolic disease, before discussing evidence for ethnic differences in disease pathophysiology between BAC and WE populations.
Collapse
Affiliation(s)
- Reuben M Reed
- Department of Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, UK
| | - Martin B Whyte
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7WG, UK
| | - Louise M Goff
- Leicester Diabetes Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
26
|
Bahramzadeh A, Bolandnazar K, Meshkani R. Resveratrol as a potential protective compound against skeletal muscle insulin resistance. Heliyon 2023; 9:e21305. [PMID: 38027557 PMCID: PMC10660041 DOI: 10.1016/j.heliyon.2023.e21305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
The increasing prevalence of type 2 diabetes has become a major global problem. Insulin resistance has a central role in pathophysiology of type 2 diabetes. Skeletal muscle is responsible for the disposal of most of the glucose under conditions of insulin stimulation, and insulin resistance in skeletal muscle causes dysregulation of glucose homeostasis in the whole body. Despite the current pharmaceutical and non-pharmacological treatment strategies to combat diabetes, there is still a need for new therapeutic agents due to the limitations of the therapeutic agents. Meanwhile, plant polyphenols have attracted the attention of researchers for their use in the treatment of diabetes and have gained popularity. Resveratrol, a stilbenoid polyphenol, exists in various plant sources, and a growing body of evidence suggests its beneficial properties, including antidiabetic activities. The present review aimed to provide a summary of the role of resveratrol in insulin resistance in skeletal muscle and its related mechanisms. To achieve the objectives, by searching the PubMed, Scopus and Web of Science databases, we have summarized the results of all cell culture, animal, and human studies that have investigated the effects of resveratrol in different models on insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Arash Bahramzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Bolandnazar
- Department of Biological Sciences and Technology, Islamic Azad University of Mashhad, Mashhad, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Musazadeh V, Tandorost A, Zarezadeh M, Jafarzadeh J, Ghavami Z, Jamilian P, Ostadrahimi A. Can omega-3 fatty acids and vitamin E co-supplementation affect obesity indices? INT J VITAM NUTR RES 2023; 93:471-480. [PMID: 35796416 DOI: 10.1024/0300-9831/a000757] [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: 11/19/2022]
Abstract
Background: Studies have shown that vitamin E as an antioxidant protects omega-3 fatty acids (FAs) from oxidation. Several studies have evaluated the effect of omega-3 FAs and vitamin E co-supplementation on obesity indices; however, the results are inconsistent. The present systematic review and meta-analysis was conducted to address the role of omega-3 FAs plus vitamin E on obesity indices. Methods: Cochrane Library, PubMed, Scopus, Embase, and Web of Science databases were searched up to February 2022. Among all of the qualified studies, 10 articles were selected. The effect size was presented as weighted mean difference (WMD) and 95% confidence interval (CI). Fixed-effects model was employed to perform meta-analysis. Subgroup analysis and publication bias assessment were carried out. Results: Ten eligible randomized controlled trials comprising 558 participants were included. The average dose of omega-3 FAs and vitamin E co-supplementation in studies was 1000-4000 mg/day and 400 IU, respectively. Intervention duration varied from 6 to 16 weeks. There was no significant effect of omega-3 and vitamin E co-supplementation on body weight (BW) (WMD=0.14 kg; 95% CI: -0.13 to 0.42; p=0.297), and body mass index (BMI) (WMD=0.08, 95% CI: -0.01 to 0.16, p=0.073). However, subgroup analysis showed that it might increase BMI in women over 50 years and if the intervention lasted more than 8 weeks. Conclusion: There was no significant impact of combined omega-3 FAs and vitamin E supplementation on BW and BMI; however, it should be noted that the intervention has an increasing impact when supplementation duration was >8 weeks and in individuals with type 2 diabetes mellitus, >50 years old, and BMI>25 kg/m2.
Collapse
Affiliation(s)
- Vali Musazadeh
- Student Research Committee, Tabriz University of Medical Sciences, Iran
- School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Iran
| | | | - Meysam Zarezadeh
- Student Research Committee, Tabriz University of Medical Sciences, Iran
- School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Iran
| | - Jaber Jafarzadeh
- School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Iran
| | - Zoha Ghavami
- Student Research Committee, Tabriz University of Medical Sciences, Iran
| | | | - Alireza Ostadrahimi
- Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Iran
| |
Collapse
|
28
|
Blaak EE, Goossens GH. Metabolic phenotyping in people living with obesity: Implications for dietary prevention. Rev Endocr Metab Disord 2023; 24:825-838. [PMID: 37581871 PMCID: PMC10492670 DOI: 10.1007/s11154-023-09830-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
Given the increasing number of people living with obesity and related chronic metabolic disease, precision nutrition approaches are required to increase the effectiveness of prevention strategies. This review addresses these approaches in different metabolic phenotypes (metabotypes) in obesity. Although obesity is typically associated with an increased cardiometabolic disease risk, some people with obesity are relatively protected against the detrimental effects of excess adiposity on cardiometabolic health, also referred to as 'metabolically healthy obesity' (MHO). Underlying mechanisms, the extent to which MHO is a transient state as well as lifestyle strategies to counteract the transition from MHO to metabolically unhealthy obesity (MUO) are discussed. Based on the limited resources that are available for dietary lifestyle interventions, it may be reasonable to prioritize interventions for people with MUO, since targeting high-risk patients for specific nutritional, lifestyle or weight-loss strategies may enhance the cost-effectiveness of these interventions. Additionally, the concept of tissue insulin resistant (IR) metabotypes is discussed, representing distinct etiologies towards type 2 diabetes (T2D) as well as cardiovascular disease (CVD). Recent evidence indicates that these tissue IR metabotypes, already present in individuals with obesity with a normal glucose homeostasis, respond differentially to diet. Modulation of dietary macronutrient composition according to these metabotypes may considerably improve cardiometabolic health benefits. Thus, nutritional or lifestyle intervention may improve cardiometabolic health, even with only minor or no weight loss, which stresses the importance of focusing on a healthy lifestyle and not on weight loss only. Targeting different metabotypes towards T2D and cardiometabolic diseases may lead to more effective lifestyle prevention and treatment strategies. Age and sex-related differences in tissue metabotypes and related microbial composition and functionality (fermentation), as important drivers and/or mediators of dietary intervention response, have to be taken into account. For the implementation of these approaches, more prospective trials are required to provide the knowledge base for precision nutrition in the prevention of chronic metabolic diseases.
Collapse
Affiliation(s)
- Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
29
|
Lempesis IG, Varrias D, Sagris M, Attaran RR, Altin ES, Bakoyiannis C, Palaiodimos L, Dalamaga M, Kokkinidis DG. Obesity and Peripheral Artery Disease: Current Evidence and Controversies. Curr Obes Rep 2023; 12:264-279. [PMID: 37243875 PMCID: PMC10220347 DOI: 10.1007/s13679-023-00510-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 05/29/2023]
Abstract
PURPOSE OF REVIEW Obesity is a significant public health problem and a major risk factor for the development and progression of atherosclerosis and its cardiovascular manifestations. Lower extremity peripheral artery disease (PAD) affects 3%-10% of the Western population and, if left untreated, can lead to devastating outcomes with both an increased risk of morbidity and mortality. Interestingly, the association between obesity and PAD remains debatable. Whereas it is well known that PAD and obesity frequently overlap in the same patients, many studies have demonstrated a negative association between obesity and PAD and a protective effect of obesity on disease development and progression, a phenomenon described as the "obesity paradox." Possible mechanisms for this paradox may include genetic background, as assessed by mendelian randomization studies, adipose tissue dysfunction, and body fat distribution rather than adiposity, while other factors, such as sex, ethnicity, sarcopenia in the elderly population, or aggressive treatment of co-existing metabolic conditions in individuals with obesity compared to those with normal weight, could have some impact as well. RECENT RINDINGS Few reviews and meta-analyses examining systematically the relationship between obesity and PAD exist. The impact of PAD development due to the presence of obesity remains largely controversial. However, the most current evidence, backed by a recent meta-analysis, suggests a potential protective role of a higher body mass index on PAD-related complications and mortality. In this review, we discuss the association between obesity and PAD development, progression, and management, and the potential pathophysiologic mechanisms linking the two diseases.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Biologic Chemistry, School of Medicine, National and Kapodistrian, University of Athens, Mikras Asias 75, 115 27, Athens, Greece.
| | - Dimitrios Varrias
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marios Sagris
- General Hospital of Nikaia, Piraeus, 184 54, Athens, Greece
| | - Robert R Attaran
- Section of Cardiovascular Medicine, Yale University/Yale New Haven Hospital, 06519, New Haven, CT, USA
| | - Elissa S Altin
- Section of Cardiovascular Medicine, Yale University/Yale New Haven Hospital, 06519, New Haven, CT, USA
| | - Christos Bakoyiannis
- Department of Surgery, Division of Vascular Surgery, Laikon General Hospital, National Kapodistrian University of Athens, 15772, Athens, Greece
| | - Leonidas Palaiodimos
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Dalamaga
- Department of Biologic Chemistry, School of Medicine, National and Kapodistrian, University of Athens, Mikras Asias 75, 115 27, Athens, Greece
| | - Damianos G Kokkinidis
- Section of Cardiovascular Medicine, Yale University/Yale New Haven Hospital, 06519, New Haven, CT, USA
| |
Collapse
|
30
|
Kim D, Memili A, Chen HH, Highland HM, Polikowsky HG, Anwar MY, Laing ST, Lee M, McCormick JB, Fisher-Hoch SP, Below JE, North KE, Gutierrez AD. Sex-specific associations between adipokine profiles and carotid-intima media thickness in the Cameron County Hispanic Cohort (CCHC). Cardiovasc Diabetol 2023; 22:231. [PMID: 37653519 PMCID: PMC10472619 DOI: 10.1186/s12933-023-01968-4] [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: 04/22/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Adipokines are hormones secreted from adipose tissue and are associated with cardiometabolic diseases (CMD). Functional differences between adipokines (leptin, adiponectin, and resistin) are known, but inconsistently reported associations with CMD and lack of studies in Hispanic populations are research gaps. We investigated the relationship between subclinical atherosclerosis and multiple adipokine measures. METHODS Cross-sectional data from the Cameron County Hispanic Cohort (N = 624; mean age = 50; Female = 70.8%) were utilized to assess associations between adipokines [continuous measures of adiponectin, leptin, resistin, leptin-to-adiponectin ratio (LAR), and adiponectin-resistin index (ARI)] and early atherosclerosis [carotid-intima media thickness (cIMT)]. We adjusted for sex, age, body mass index (BMI), smoking status, cytokines, fasting blood glucose levels, blood pressure, lipid levels, and medication usage in the fully adjusted linear regression model. We conducted sexes-combined and sex-stratified analyses to account for sex-specificity and additionally tested whether stratification of participants by their metabolic status (metabolically elevated risk for CMD as defined by having two or more of the following conditions: hypertension, dyslipidemia, insulin resistance, and inflammation vs. not) influenced the relationship between adipokines and cIMT. RESULTS In the fully adjusted analyses, adiponectin, leptin, and LAR displayed significant interaction by sex (p < 0.1). Male-specific associations were between cIMT and LAR [β(SE) = 0.060 (0.016), p = 2.52 × 10-4], and female-specific associations were between cIMT and adiponectin [β(SE) = 0.010 (0.005), p = 0.043] and ARI [β(SE) = - 0.011 (0.005), p = 0.036]. When stratified by metabolic health status, the male-specific positive association between LAR and cIMT was more evident among the metabolically healthy group [β(SE) = 0.127 (0.015), p = 4.70 × 10-10] (p for interaction by metabolic health < 0.1). However, the female-specific associations between adiponectin and cIMT and ARI and cIMT were observed only among the metabolically elevated risk group [β(SE) = 0.014 (0.005), p = 0.012 for adiponectin; β(SE) = - 0.015 (0.006), p = 0.013 for ARI; p for interaction by metabolic health < 0.1]. CONCLUSION Associations between adipokines and cIMT were sex-specific, and metabolic health status influenced the relationships between adipokines and cIMT. These heterogeneities by sex and metabolic health affirm the complex relationships between adipokines and atherosclerosis.
Collapse
Affiliation(s)
- Daeeun Kim
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aylin Memili
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hannah G Polikowsky
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Yaser Anwar
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Susan T Laing
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Joseph B McCormick
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Absalon D Gutierrez
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
31
|
Keshavjee SH, Schwenger KJP, Yadav J, Pickel L, Ghorbani Y, Sung HK, Jung H, Lou W, Fischer SE, Jackson TD, Okrainec A, Allard JP. Adipose Tissue and Plasma Markers Associated with HbA1c Pre- and Post-bariatric Surgery: a Cross-sectional and Cohort Study. Obes Surg 2023; 33:2443-2451. [PMID: 37380880 DOI: 10.1007/s11695-023-06679-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023]
Abstract
PURPOSE Obesity can be associated with chronic inflammation and dysregulated expression of inflammatory adipokines that contribute to insulin resistance and type 2 diabetes. This may also affect the clinical response to bariatric surgery. Our objective was whether baseline visceral adipose tissue features and plasma adipokine are associated with HbA1c ≥0.06 at the time of Roux-en-Y gastric bypass (RYGB) surgery and with persistently elevated HbA1c at 12 months post-RYGB. METHODS During the surgery, adipose biopsies and plasma were collected for adipokine/cytokine profile. Clinical and biochemical measurements were also collected at the time of RYGB and, in those with baseline elevated HbA1c, at 12 months post-RYGB. RESULTS In the cross-sectional study, 109 patients (82.6% female; age 49 years; BMI 46.98 kg/m2) participated. Of those with elevated HbA1c at baseline (n = 61), 47 patients had repeated measurements at 12 months post-RYGB (23% drop-out). Using a multivariate logistic regression model, older age (adjusted odds ratio (aOR), 1.14; 95% confidence interval (CI), 1.06-1.22) and higher plasma resistin (aOR, 5.30; 95% CI, 1.25-22.44) were associated with higher odds of HbA1c ≥ 0.06, whereas higher plasma adiponectin (aOR, 0.993; 95% CI, 0.99-0.996) was associated with lower odds of HbA1c ≥0.06. In addition, baseline higher average adipose cell area (aOR, 1.0017; 95% CI, 1.0002-1.0032) and plasma resistin (aOR, 1.0004; 95% CI, 1.0000-1.0009) were associated with higher odds of having persistently elevated HbA1c at 12 months post-RYGB. CONCLUSION Our study suggests that baseline plasma adipokine dysregulation, specifically high resistin, and adipocyte hypertrophy may affect the clinical response to RYGB.
Collapse
Affiliation(s)
- Sara H Keshavjee
- Division of General Surgery, University Health Network, University of Toronto, 190 Elizabeth Street, 1st Floor, Suite 408, Toronto, ON, M5G 2C4, Canada
| | - Katherine J P Schwenger
- Division of Gastroenterology, Toronto General Hospital, University Health Network, 6 Queen's Park Crescent West, Third Floor, Toronto, ON, M5S 3H2, Canada
| | - Jitender Yadav
- Department of Immunology, University of Toronto, 1 King's College Circle, Medical Sciences Building, Room 7207, Toronto, ON, M5S 1A8, Canada
| | - Lauren Pickel
- Translational Medicine Program, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
| | - Yasaman Ghorbani
- Division of Gastroenterology, Toronto General Hospital, University Health Network, 6 Queen's Park Crescent West, Third Floor, Toronto, ON, M5S 3H2, Canada
| | - Hoon-Ki Sung
- Translational Medicine Program, The Hospital for Sick Children, 686 Bay St, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, 6th Floor, Toronto, ON, M5S 1A8, Canada
| | - Hyejung Jung
- Dalla Lana Public Health Department, University of Toronto, 155 College St, 6th Fl, Toronto, ON, M5T 3M7, Canada
| | - Wendy Lou
- Dalla Lana Public Health Department, University of Toronto, 155 College St, 6th Fl, Toronto, ON, M5T 3M7, Canada
| | - Sandra E Fischer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, 6th Floor, Toronto, ON, M5S 1A8, Canada
| | - Timothy D Jackson
- Division of General Surgery, University Health Network, University of Toronto, 190 Elizabeth Street, 1st Floor, Suite 408, Toronto, ON, M5G 2C4, Canada
| | - Allan Okrainec
- Division of General Surgery, University Health Network, University of Toronto, 190 Elizabeth Street, 1st Floor, Suite 408, Toronto, ON, M5G 2C4, Canada
| | - Johane P Allard
- Division of Gastroenterology, Toronto General Hospital, University Health Network, 6 Queen's Park Crescent West, Third Floor, Toronto, ON, M5S 3H2, Canada.
| |
Collapse
|
32
|
Erdős B, van Sloun B, Goossens GH, O'Donovan SD, de Galan BE, van Greevenbroek MMJ, Stehouwer CDA, Schram MT, Blaak EE, Adriaens ME, van Riel NAW, Arts ICW. Quantifying postprandial glucose responses using a hybrid modeling approach: Combining mechanistic and data-driven models in The Maastricht Study. PLoS One 2023; 18:e0285820. [PMID: 37498860 PMCID: PMC10374070 DOI: 10.1371/journal.pone.0285820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 05/03/2023] [Indexed: 07/29/2023] Open
Abstract
Computational models of human glucose homeostasis can provide insight into the physiological processes underlying the observed inter-individual variability in glucose regulation. Modelling approaches ranging from "bottom-up" mechanistic models to "top-down" data-driven techniques have been applied to untangle the complex interactions underlying progressive disturbances in glucose homeostasis. While both approaches offer distinct benefits, a combined approach taking the best of both worlds has yet to be explored. Here, we propose a sequential combination of a mechanistic and a data-driven modeling approach to quantify individuals' glucose and insulin responses to an oral glucose tolerance test, using cross sectional data from 2968 individuals from a large observational prospective population-based cohort, the Maastricht Study. The best predictive performance, measured by R2 and mean squared error of prediction, was achieved with personalized mechanistic models alone. The addition of a data-driven model did not improve predictive performance. The personalized mechanistic models consistently outperformed the data-driven and the combined model approaches, demonstrating the strength and suitability of bottom-up mechanistic models in describing the dynamic glucose and insulin response to oral glucose tolerance tests.
Collapse
Affiliation(s)
- Balázs Erdős
- TiFN, Wageningen, Netherlands
- MaCSBio Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Bart van Sloun
- TiFN, Wageningen, Netherlands
- MaCSBio Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Gijs H Goossens
- TiFN, Wageningen, Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Shauna D O'Donovan
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Bastiaan E de Galan
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marleen M J van Greevenbroek
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Coen D A Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Miranda T Schram
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
- MHeNs School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| | - Ellen E Blaak
- TiFN, Wageningen, Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michiel E Adriaens
- TiFN, Wageningen, Netherlands
- MaCSBio Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Natal A W van Riel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Ilja C W Arts
- MaCSBio Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
33
|
Lempesis IG, Hoebers N, Essers Y, Jocken JWE, Dineen R, Blaak EE, Manolopoulos KN, Goossens GH. Distinct inflammatory signatures of upper and lower body adipose tissue and adipocytes in women with normal weight or obesity. Front Endocrinol (Lausanne) 2023; 14:1205799. [PMID: 37455922 PMCID: PMC10338223 DOI: 10.3389/fendo.2023.1205799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Upper and lower body fat accumulation poses an opposing obesity-related cardiometabolic disease risk. Depot-differences in subcutaneous adipose tissue (SAT) function may underlie these associations. We aimed to investigate the inflammatory signatures of abdominal (ABD) and femoral (FEM) SAT in postmenopausal women with normal weight or obesity. Methods We included 23 postmenopausal women with normal weight (n = 13) or obesity (n = 10). In vivo secretion of adipokines from ABD and FEM SAT was measured using the arterio-venous balance technique. Adipokine gene expression and adipocyte morphology were examined in ABD and FEM SAT. Furthermore, adipokine expression and secretion were investigated in vitro using differentiated human primary ABD and FEM subcutaneous adipocytes derived from the study participants. Results Plasma leptin and plasminogen activator inhibitor (PAI)-1 concentrations were higher, and ABD and FEM adipocytes were larger in women with obesity than normal weight. No differences in adipocyte size and blood flow were apparent between ABD and FEM SAT. We found significant release of leptin and monocyte chemoattractant protein (MCP)-1 from ABD and FEM SAT, with higher fractional release of MCP-1 from ABD than FEM SAT. Gene expression of leptin, PAI-1, and tumor necrosis factor-α was lower in ABD than FEM SAT and higher in women with obesity than normal weight. In ABD adipocytes, interleukin-6, PAI-1, and leptin gene expression were higher, while adiponectin and dipeptidyl-peptidase-4 gene expression were lower than in FEM adipocytes. Finally, ABD adipocytes secreted less MCP-1 compared to FEM adipocytes. Discussion These findings demonstrate that upper and lower body SAT and adipocytes are characterized by distinct inflammatory signatures in postmenopausal women, which seem independent of adipocyte size.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Johan W. E. Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Rosemary Dineen
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| | - Konstantinos N. Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands, Maastricht, Netherlands
| |
Collapse
|
34
|
Shaik Mohamed Sayed UF, Moshawih S, Goh HP, Kifli N, Gupta G, Singh SK, Chellappan DK, Dua K, Hermansyah A, Ser HL, Ming LC, Goh BH. Natural products as novel anti-obesity agents: insights into mechanisms of action and potential for therapeutic management. Front Pharmacol 2023; 14:1182937. [PMID: 37408757 PMCID: PMC10318930 DOI: 10.3389/fphar.2023.1182937] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 07/07/2023] Open
Abstract
Obesity affects more than 10% of the adult population globally. Despite the introduction of diverse medications aimed at combating fat accumulation and obesity, a significant number of these pharmaceutical interventions are linked to substantial occurrences of severe adverse events, occasionally leading to their withdrawal from the market. Natural products serve as attractive sources for anti-obesity agents as many of them can alter the host metabolic processes and maintain glucose homeostasis via metabolic and thermogenic stimulation, appetite regulation, pancreatic lipase and amylase inhibition, insulin sensitivity enhancing, adipogenesis inhibition and adipocyte apoptosis induction. In this review, we shed light on the biological processes that control energy balance and thermogenesis as well as metabolic pathways in white adipose tissue browning, we also highlight the anti-obesity potential of natural products with their mechanism of action. Based on previous findings, the crucial proteins and molecular pathways involved in adipose tissue browning and lipolysis induction are uncoupling protein-1, PR domain containing 16, and peroxisome proliferator-activated receptor-γ in addition to Sirtuin-1 and AMP-activated protein kinase pathway. Given that some phytochemicals can also lower proinflammatory substances like TNF-α, IL-6, and IL-1 secreted from adipose tissue and change the production of adipokines like leptin and adiponectin, which are important regulators of body weight, natural products represent a treasure trove for anti-obesity agents. In conclusion, conducting comprehensive research on natural products holds the potential to accelerate the development of an improved obesity management strategy characterized by heightened efficacy and reduced incidence of side effects.
Collapse
Affiliation(s)
| | - Said Moshawih
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Hui Poh Goh
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Nurolaini Kifli
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Andi Hermansyah
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
| | - Hooi Leng Ser
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas AirlanggaSurabaya, Indonesia
- School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
35
|
Lempesis IG, Georgakopoulou VE. Physiopathological mechanisms related to inflammation in obesity and type 2 diabetes mellitus. World J Exp Med 2023; 13:7-16. [PMID: 37396883 PMCID: PMC10308320 DOI: 10.5493/wjem.v13.i3.7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/15/2023] [Accepted: 04/10/2023] [Indexed: 06/16/2023] Open
Abstract
Overweight, obesity, and type 2 diabetes mellitus pose global health problems that are ever-increasing. A chronic low-grade inflammatory status and the presence of various pro-inflammatory markers either in circulation or within dysfunctional metabolic tissues are well established. The presence of these factors can, to some extent, predict disease development and progression. A central role is played by the presence of dysfunctional adipose tissue, liver dysfunction, and skeletal muscle dysfunction, which collectively contribute to the increased circulatory levels of proinflammatory factors. Weight loss and classical metabolic interventions achieve a decrease in many of these factors' circulating levels, implying that a better understanding of the processes or even the modulation of inflammation may alleviate these diseases. This review suggests that inflammation plays a significant role in the development and progression of these conditions and that measuring inflammatory markers may be useful for assessing disease risk and development of future treatment methods.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Athens 11527, Greece
| | | |
Collapse
|
36
|
Cuciureanu M, Caratașu CC, Gabrielian L, Frăsinariu OE, Checheriță LE, Trandafir LM, Stanciu GD, Szilagyi A, Pogonea I, Bordeianu G, Soroceanu RP, Andrițoiu CV, Anghel MM, Munteanu D, Cernescu IT, Tamba BI. 360-Degree Perspectives on Obesity. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1119. [PMID: 37374323 PMCID: PMC10304508 DOI: 10.3390/medicina59061119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023]
Abstract
Alarming statistics show that the number of people affected by excessive weight has surpassed 2 billion, representing approximately 30% of the world's population. The aim of this review is to provide a comprehensive overview of one of the most serious public health problems, considering that obesity requires an integrative approach that takes into account its complex etiology, including genetic, environmental, and lifestyle factors. Only an understanding of the connections between the many contributors to obesity and the synergy between treatment interventions can ensure satisfactory outcomes in reducing obesity. Mechanisms such as oxidative stress, chronic inflammation, and dysbiosis play a crucial role in the pathogenesis of obesity and its associated complications. Compounding factors such as the deleterious effects of stress, the novel challenge posed by the obesogenic digital (food) environment, and the stigma associated with obesity should not be overlooked. Preclinical research in animal models has been instrumental in elucidating these mechanisms, and translation into clinical practice has provided promising therapeutic options, including epigenetic approaches, pharmacotherapy, and bariatric surgery. However, more studies are necessary to discover new compounds that target key metabolic pathways, innovative ways to deliver the drugs, the optimal combinations of lifestyle interventions with allopathic treatments, and, last but not least, emerging biological markers for effective monitoring. With each passing day, the obesity crisis tightens its grip, threatening not only individual lives but also burdening healthcare systems and societies at large. It is high time we took action as we confront the urgent imperative to address this escalating global health challenge head-on.
Collapse
Affiliation(s)
- Magdalena Cuciureanu
- Department of Pharmacology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (C.-C.C.); (I.T.C.); (B.I.T.)
| | - Cătălin-Cezar Caratașu
- Department of Pharmacology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (C.-C.C.); (I.T.C.); (B.I.T.)
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Levon Gabrielian
- Department of Anatomy and Pathology, The University of Adelaide, Adelaide 5000, Australia;
| | - Otilia Elena Frăsinariu
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Laura Elisabeta Checheriță
- 2nd Dental Medicine Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Mihaela Trandafir
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Andrei Szilagyi
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| | - Ina Pogonea
- Department of Pharmacology and Clinical Pharmacology, “Nicolae Testemiţanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova; (I.P.); (M.M.A.)
| | - Gabriela Bordeianu
- Department of Biochemistry, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Radu Petru Soroceanu
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Călin Vasile Andrițoiu
- Specialization of Nutrition and Dietetics, “Vasile Goldis” Western University of Arad, 310025 Arad, Romania
| | - Maria Mihalache Anghel
- Department of Pharmacology and Clinical Pharmacology, “Nicolae Testemiţanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova; (I.P.); (M.M.A.)
| | - Diana Munteanu
- Institute of Mother and Child, “Nicolae Testemiţanu” State University of Medicine and Pharmacy, 2062 Chisinau, Moldova;
| | - Irina Teodora Cernescu
- Department of Pharmacology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (C.-C.C.); (I.T.C.); (B.I.T.)
| | - Bogdan Ionel Tamba
- Department of Pharmacology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (C.-C.C.); (I.T.C.); (B.I.T.)
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (A.S.)
| |
Collapse
|
37
|
Gruzdeva O, Dyleva Y, Belik E, Uchasova E, Ponasenko A, Ivanov S, Zinets M, Stasev A, Kutikhin A, Markova V, Poddubnyak A, Gorbatovskaya E, Fanaskova E, Barbarash O. Expression of Ceramide-Metabolizing Enzymes in the Heart Adipose Tissue of Cardiovascular Disease Patients. Int J Mol Sci 2023; 24:ijms24119494. [PMID: 37298446 DOI: 10.3390/ijms24119494] [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: 05/04/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Here, we examined the expression of ceramide metabolism enzymes in the subcutaneous adipose tissue (SAT), epicardial adipose tissue (EAT) and perivascular adipose tissue (PVAT) of 30 patients with coronary artery disease (CAD) and 30 patients with valvular heart disease (VHD) by means of quantitative polymerase chain reaction and fluorescent Western blotting. The EAT of patients with CAD showed higher expression of the genes responsible for ceramide biosynthesis (SPTLC1, SPTLC2, CERS1, 5, 6, DEGS1, and SMPD1) and utilization (ASAH1, SGMS1). PVAT was characterized by higher mRNA levels of CERS3, CERS4, DEGS1, SMPD1, and ceramide utilization enzyme (SGMS2). In patients with VHD, there was a high CERS4, DEGS1, and SGMS2 expression in the EAT and CERS3 and CERS4 expression in the PVAT. Among patients with CAD, the expression of SPTLC1 in SAT and EAT, SPTLC2 in EAT, CERS2 in all studied AT, CERS4 and CERS5 in EAT, DEGS1 in SAT and EAT, ASAH1 in all studied AT, and SGMS1 in EAT was higher than in those with VHD. Protein levels of ceramide-metabolizing enzymes were consistent with gene expression trends. The obtained results indicate an activation of ceramide synthesis de novo and from sphingomyelin in cardiovascular disease, mainly in EAT, that contributes to the accumulation of ceramides in this location.
Collapse
Affiliation(s)
- Olga Gruzdeva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
- Department of Pathophysiology, Kemerovo State Medical University, 650029 Kemerovo, Russia
| | - Yulia Dyleva
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Ekaterina Belik
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Evgenia Uchasova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anastasia Ponasenko
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Sergey Ivanov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Maxim Zinets
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Alexander Stasev
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Alena Poddubnyak
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Evgenia Gorbatovskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Elena Fanaskova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
| | - Olga Barbarash
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnovy Boulevard, 650002 Kemerovo, Russia
- Department of Pathophysiology, Kemerovo State Medical University, 650029 Kemerovo, Russia
| |
Collapse
|
38
|
Lempesis IG, Georgakopoulou VE. Implications of obesity and adiposopathy on respiratory infections; focus on emerging challenges. World J Clin Cases 2023; 11:2925-2933. [PMID: 37215426 PMCID: PMC10198078 DOI: 10.12998/wjcc.v11.i13.2925] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Obesity is characterized by excessive adipose tissue accumulation, which impacts physiological, metabolic, and immune functions. Several respiratory infections, including bacterial pneumonia, influenza, and coronavirus disease 2019, appear to be linked to unfavorable results in individuals with obesity. These may be attributed to the direct mechanical/physiological effects of excess body fat on the lungs’ function. Notably, adipose tissue dysfunction is associated with a low-grade chronic inflammatory status and hyperleptinemia, among other characteristics. These have all been linked to immune system dysfunction and weakened immune responses to these infections. A better understanding and clinical awareness of these risk factors are necessary for better disease outcomes.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Athens 11527, Greece
| | | |
Collapse
|
39
|
Lempesis IG, Karlafti E, Papalexis P, Fotakopoulos G, Tarantinos K, Lekakis V, Papadakos SP, Cholongitas E, Georgakopoulou VE. COVID-19 and liver injury in individuals with obesity. World J Gastroenterol 2023; 29:908-916. [PMID: 36844135 PMCID: PMC9950870 DOI: 10.3748/wjg.v29.i6.908] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
Coronavirus disease 2019 is an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 that manifests as a variety of clinical manifestations, including liver damage commonly detected by a hepatocellular pattern from liver function tests. Liver injury is associated with a worse prognosis overall. Conditions associated with the severity of the disease include obesity and cardiometabolic comorbidities, which are also associated with nonalcoholic fatty liver disease (NAFLD). The presence of NAFLD, similarly to obesity, is associated with an unfavourable impact on the coronavirus disease 2019 outcome. Individuals with these conditions could present with liver damage and elevated liver function tests due to direct viral cytotoxicity, systemic inflammation, ischemic or hypoxic liver damage or drug side effects. However, liver damage in the setting of NAFLD could also be attributed to a pre-existing chronic low-grade inflammation associated with surplus and dysfunctional adipose tissue in these individuals. Here we investigate the hypothesis that a pre-existing inflammatory status is exacerbated after severe acute respiratory syndrome coronavirus 2 infection, which embodies a second hit to the underestimated liver damage.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, United Kingdom
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht 616 6200, Netherlands
| | - Eleni Karlafti
- Department of Emergency, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki 546 21, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
- Department of Biomedical Sciences, University of West Attica, Athens 12243, Greece
| | - George Fotakopoulos
- Department of Neurosurgery, General University Hospital of Larisa, Larisa 41221, Greece
| | | | - Vasileios Lekakis
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Stavros P Papadakos
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | | |
Collapse
|
40
|
Dietary n-3 and n-6 polyunsaturated fatty acids differentially modulate the adiponectin and leptinmediated major signaling pathways in visceral and subcutaneous white adipose tissue in high fat diet induced obesity in Wistar rats. Nutr Res 2023; 110:74-86. [PMID: 36689814 DOI: 10.1016/j.nutres.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 12/12/2022] [Accepted: 12/18/2022] [Indexed: 12/29/2022]
Abstract
Obesity is a chronic metabolic disease that involves excessive accumulation of fat in white adipose tissue (WAT). Apart from storing excess fats, WAT also serves as an important endocrine organ secreting adipocytokines such as adiponectin and leptin. Adiponectin and leptin bind to their transmembrane receptors adiponectin receptor 1 (AdipoR1)/adiponectin receptor 2 (AdipoR2) and Ob-R, respectively, and mediate their effect on metabolism by regulating multiple downstream targets. Dietary fat is considered the main culprit behind obesity development. Numerous preclinical studies have highlighted role of essential polyunsaturated fatty acids (PUFAs), particularly n-3 PUFAs, in prevention of obesity. Despite emerging data, there still is no clear understanding of the mechanism of action of n-3 PUFAs and n-6 PUFAs on adipose tissue function in two functionally and anatomically different depots of WAT: visceral and subcutaneous. We designed this study using a high fat diet (HFD) fed rodent model of obesity to test our hypothesis that n-3 and n-6 PUFAs possibly differentially modulate adipokine secretion and downstream metabolic pathways such as peroxisome proliferator-activated receptor-γ (PPAR-γ), protein kinase B (AKT)-forkhead box O1 (FOXO1), and Janus kinase-signal transducer and activator of transcription in obesity. The results of the current study showed that n-3 PUFAs upregulate the expression of AdipoR1/R2 and ameliorate the effects of HFD by modulating adipogenesis via PPAR-γ and by improving glucose tolerance and lipid metabolism via AKT-FOXO1 axis in fish oil fed rats. However, n-6 PUFAs did not show any remarkable change compared with HFD fed animals. Our study highlights that n-3 PUFAs modulate expression of various targets in adiponectin and leptin signaling cascade, bringing about an overall reduction in obesity and improvement in adipose tissue function in HFD induced obesity.
Collapse
|
41
|
Akash MSH, Rasheed S, Rehman K, Ibrahim M, Imran M, Assiri MA. Biochemical Activation and Regulatory Functions of Trans-Regulatory KLF14 and Its Association with Genetic Polymorphisms. Metabolites 2023; 13:metabo13020199. [PMID: 36837818 PMCID: PMC9962810 DOI: 10.3390/metabo13020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Krüpple-Like family of transcription factor-14 (KLF14) is a master trans-regulatory gene that has multiple biological regulatory functions and is involved in many pathological mechanisms. It controls the expressions of several other genes which are involved in multiple regulatory functions. KLF14 plays a significant role in lipid metabolism, glucose regulation and insulin sensitivity. Cell apoptosis, proliferation, and differentiation are regulated by the KLF14 gene, and up-regulation of KLF14 prevents cancer progression. KLF14 has been used as an epigenetic biomarker for the estimation of chronological age due to the presence of different age-related CpG sites on genes that become methylated with age. Different genome-wide association studies have identified several KLF14 variants in adipose tissues. These single nucleotide polymorphisms in KLF14 have been associated with dyslipidemia, insulin resistance, and glucose intolerance. Moreover, the prevalence of genetic polymorphism is different in different populations due to ethnic differences and epigenetic modifications. In addition, environmental and physiological factors such as diet, age, gender, and obesity are also responsible for genetic mutations in KLF14.
Collapse
Affiliation(s)
- Muhammad Sajid Hamid Akash
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.S.H.A.); (K.R.)
| | - Sumbal Rasheed
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan 60000, Pakistan
- Correspondence: (M.S.H.A.); (K.R.)
| | - Muhammad Ibrahim
- Department of Applied Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Imran
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha 62413, Saudi Arabia
| | - Mohammed A. Assiri
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 62413, Saudi Arabia
- Department of Chemistry, Faculty of Science, King Khalid University, Abha 62413, Saudi Arabia
| |
Collapse
|
42
|
Kassis A, Fichot MC, Horcajada MN, Horstman AMH, Duncan P, Bergonzelli G, Preitner N, Zimmermann D, Bosco N, Vidal K, Donato-Capel L. Nutritional and lifestyle management of the aging journey: A narrative review. Front Nutr 2023; 9:1087505. [PMID: 36761987 PMCID: PMC9903079 DOI: 10.3389/fnut.2022.1087505] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
With age, the physiological responses to occasional or regular stressors from a broad range of functions tend to change and adjust at a different pace and restoring these functions in the normal healthy range becomes increasingly challenging. Even if this natural decline is somehow unavoidable, opportunities exist to slow down and attenuate the impact of advancing age on major physiological processes which, when weakened, constitute the hallmarks of aging. This narrative review revisits the current knowledge related to the aging process and its impact on key metabolic functions including immune, digestive, nervous, musculoskeletal, and cardiovascular functions; and revisits insights into the important biological targets that could inspire effective strategies to promote healthy aging.
Collapse
Affiliation(s)
- Amira Kassis
- Whiteboard Nutrition Science, Beaconsfield, QC, Canada,Amira Kassis,
| | | | | | | | - Peter Duncan
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | | | - Nicolas Preitner
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Diane Zimmermann
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Karine Vidal
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Laurence Donato-Capel
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland,*Correspondence: Laurence Donato-Capel,
| |
Collapse
|
43
|
Marsh ML, Oliveira MN, Vieira-Potter VJ. Adipocyte Metabolism and Health after the Menopause: The Role of Exercise. Nutrients 2023; 15:444. [PMID: 36678314 PMCID: PMC9862030 DOI: 10.3390/nu15020444] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Postmenopausal women represent an important target population in need of preventative cardiometabolic approaches. The loss of estrogen following the menopause eliminates protections against metabolic dysfunction, largely due to its role in the health and function of adipose tissue. In addition, some studies associate the menopause with reduced physical activity, which could potentially exacerbate the deleterious cardiometabolic risk profile accompanying the menopause. Meanwhile, exercise has adipocyte-specific effects that may alleviate the adverse impact of estrogen loss through the menopausal transition period and beyond. Exercise thus remains the best therapeutic agent available to mitigate menopause-associated metabolic dysfunction and represents a vital behavioral strategy to prevent and alleviate health decline in this population.
Collapse
|
44
|
Drabińska N, Romaszko J, White P. The effect of isocaloric, energy-restrictive, KETOgenic diet on metabolism, inflammation, nutrition deficiencies and oxidative stress in women with overweight and obesity (KETO-MINOX): Study protocol. PLoS One 2023; 18:e0285283. [PMID: 37155645 PMCID: PMC10166534 DOI: 10.1371/journal.pone.0285283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/12/2023] [Indexed: 05/10/2023] Open
Abstract
Obesity is considered one of the biggest health problems of the 21st century, becoming a worldwide epidemic, leading to the development of many diseases and increasing the risk of premature death. The first step in reducing body weight is a calorie-restricted diet. To date, there are many different diet types available, including the ketogenic diet (KD) which is recently gaining a lot of attention. However, all the physiological consequences of KD in the human body are not fully understood. Therefore, this study aims to evaluate the effectiveness of an eight-week, isocaloric, energy-restricted, KD as a weight management solution in women with overweight and obesity compared to a standard, balanced diet with the same calorie content. The primary outcome is to evaluate the effects of a KD on body weight and composition. The secondary outcomes are to evaluate the effect of KD-related weight loss on inflammation, oxidative stress, nutritional status, profiles of metabolites in breath, which informs about the metabolic changes in the body, obesity and diabetes-associated parameters, including a lipid profile, status of adipokines and hormones. Notably, in this trial, the long-term effects and efficiency of the KD will be studied. In summary, the proposed study will fill the gap in knowledge about the effects of KD on inflammation, obesity-associated parameters, nutritional deficiencies, oxidative stress and metabolism in a single study. ClinicalTrail.gov registration number: NCT05652972.
Collapse
Affiliation(s)
- Natalia Drabińska
- Department of Chemistry and Biodynamics of Food, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Jerzy Romaszko
- Department of Family Medicine and Infectious Diseases, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Paul White
- Department of Mathematics and Data Science, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
45
|
Gomes MM, da Silva MMR, de Araújo IM, de Paula FJA. Bone, fat, and muscle interactions in health and disease. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2022; 66:611-620. [PMID: 36382750 PMCID: PMC10118823 DOI: 10.20945/2359-3997000000550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022]
Abstract
Energy metabolism is a point of integration among the various organs and tissues of the human body, not only in terms of consumption of energy substrates but also because it concentrates a wide interconnected network controlled by endocrine factors. Thus, not only do tissues consume substrates, but they also participate in modulating energy metabolism. Soft mesenchymal tissues, in particular, play a key role in this process. The recognition that high energy consumption is involved in bone remodeling has been accompanied by evidence showing that osteoblasts and osteocytes produce factors that influence, for example, insulin sensitivity and appetite. Additionally, there are significant interactions between muscle, adipose, and bone tissues to control mutual tissue trophism. Not by chance, trophic and functional changes in these tissues go hand in hand from the beginning of an individual's development until aging. Likewise, metabolic and nutritional diseases deeply affect the musculoskeletal system and adipose tissue. The present narrative review highlights the importance of the interaction of the mesenchymal tissues for bone development and maintenance and the impact on bone from diseases marked by functional and trophic disorders of adipose and muscle tissues.
Collapse
Affiliation(s)
- Mayra Macena Gomes
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | - Iana Mizumukai de Araújo
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, SP, Brasil
| | | |
Collapse
|
46
|
Lempesis IG, Hoebers N, Essers Y, Jocken JWE, Rouschop KMA, Blaak EE, Manolopoulos KN, Goossens GH. Physiological Oxygen Levels Differentially Regulate Adipokine Production in Abdominal and Femoral Adipocytes from Individuals with Obesity Versus Normal Weight. Cells 2022; 11:cells11223532. [PMID: 36428961 PMCID: PMC9688196 DOI: 10.3390/cells11223532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue (AT) inflammation may increase obesity-related cardiometabolic complications. Altered AT oxygen partial pressure (pO2) may impact the adipocyte inflammatory phenotype. Here, we investigated the effects of physiological pO2 levels on the inflammatory phenotype of abdominal (ABD) and femoral (FEM) adipocytes derived from postmenopausal women with normal weight (NW) or obesity (OB). Biopsies were collected from ABD and FEM subcutaneous AT in eighteen postmenopausal women (aged 50-65 years) with NW (BMI 18-25 kg/m2, n = 9) or OB (BMI 30-40 kg/m2, n = 9). We compared the effects of prolonged exposure to different physiological pO2 levels on adipokine expression and secretion in differentiated human multipotent adipose-derived stem cells. Low physiological pO2 (5% O2) significantly increased leptin gene expression/secretion in ABD and FEM adipocytes derived from individuals with NW and OB compared with high physiological pO2 (10% O2) and standard laboratory conditions (21% O2). Gene expression/secretion of IL-6, DPP-4, and MCP-1 was reduced in differentiated ABD and FEM adipocytes from individuals with OB but not NW following exposure to low compared with high physiological pO2 levels. Low physiological pO2 decreases gene expression and secretion of several proinflammatory factors in ABD and FEM adipocytes derived from individuals with OB but not NW.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
- Correspondence: (I.G.L.); (G.H.G.)
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Johan W. E. Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Kasper M. A. Rouschop
- Radiotherapy, GROW School for Oncology & Reproduction, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Konstantinos N. Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Correspondence: (I.G.L.); (G.H.G.)
| |
Collapse
|
47
|
Li Y, Wang H, Chen H, Liao Y, Gou S, Yan Q, Zhuang Z, Li H, Wang J, Suo Y, Lan T, Liu Y, Zhao Y, Zou Q, Nie T, Hui X, Lai L, Wu D, Fan N. Generation of a genetically modified pig model with CREBRF R457Q variant. FASEB J 2022; 36:e22611. [PMID: 36250915 DOI: 10.1096/fj.202201117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
Obesity is among the strongest risk factors for type 2 diabetes (T2D). The CREBRF missense allele rs373863828 (p. Arg457Gln, p. R457Q) is associated with increased body mass index but reduced risk of T2D in people of Pacific ancestry. To investigate the functional consequences of the CREBRF variant, we introduced the corresponding human mutation R457Q into the porcine genome. The CREBRFR457Q pigs displayed dramatically increased fat deposition, which was mainly distributed in subcutaneous adipose tissue other than visceral adipose tissue. The CREBRFR457Q variant promoted preadipocyte differentiation. The increased differentiation capacity of precursor adipocytes conferred pigs the unique histological phenotype that adipocytes had a smaller size but a greater number in subcutaneous adipose tissue (SAT) of CREBRFR457Q variant pigs. In addition, in SAT of CREBRFR457Q pigs, the contents of the peroxidative metabolites 4-hydroxy-nonenal and malondialdehyde were significantly decreased, while the activity of antioxidant enzymes, such as glutathione peroxidase, superoxide dismutase, and catalase, was increased, which was in accordance with the declined level of the reactive oxygen species (ROS) in CREBRFR457Q pigs. Together, these data supported a causal role of the CREBRFR457Q variant in the pathogenesis of obesity, partly via adipocyte hyperplasia, and further suggested that reduced oxidative stress in adipose tissue may mediate the relative metabolic protection afforded by this variant despite the related obesity.
Collapse
Affiliation(s)
- Yingying Li
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Chinese Academy of Sciences, Beijing, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hai Wang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Huangyao Chen
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yuan Liao
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Shixue Gou
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Quanmei Yan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhenpeng Zhuang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Hao Li
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Jiaowei Wang
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yangyang Suo
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Ting Lan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yang Liu
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yu Zhao
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Qingjian Zou
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Tao Nie
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoyan Hui
- School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong SAR
| | - Liangxue Lai
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China.,Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, China
| | - Donghai Wu
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Nana Fan
- CAS Key Laboratory of Regenerative Biology, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Sanya Institute of Swine Resource, Hainan Provincial Research Centre of Laboratory Animals, Sanya, China.,Research Unit of Generation of Large Animal Disease Models, Chinese Academy of Medical Sciences (2019RU015), Guangzhou, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| |
Collapse
|
48
|
Glycerol-3-phosphate Acyltransferases and Metabolic Syndrome: Recent Advances and Future Perspectives. Expert Rev Mol Med 2022; 24:e30. [PMID: 36059117 DOI: 10.1017/erm.2022.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
49
|
Of mice and men: Considerations on adipose tissue physiology in animal models of obesity and human studies. Metabol Open 2022; 15:100208. [PMID: 36092796 PMCID: PMC9460138 DOI: 10.1016/j.metop.2022.100208] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 12/26/2022] Open
Abstract
The ever-increasing burden of obesity demands a better pathophysiological understanding, especially regarding adipose tissue pathophysiology. Animal models of obesity are of great importance in investigating potential mechanisms and implications of obesity. Many issues should be considered while interpreting the preclinical results as anatomical and pathophysiological differences exist among species. Importantly, the natural history of obesity development differs considerably. An important example of conflicting results among preclinical models and human physiological studies is that of adipose tissue oxygenation, where rodent models almost unanimously have shown the presence of hypoxia in the adipose tissue of obese animals while human studies have yielded conflicting results to date. Other issues which require further clarification before generalizing preclinical data in humans include adipose tissue browning, endocrine function and fibrosis. The aim of this mini-review is to synopsize similarities and differences between rodent models and humans, which should be taken into consideration in obesity studies.
Collapse
|
50
|
Vanweert F, Schrauwen P, Phielix E. Role of branched-chain amino acid metabolism in the pathogenesis of obesity and type 2 diabetes-related metabolic disturbances BCAA metabolism in type 2 diabetes. Nutr Diabetes 2022; 12:35. [PMID: 35931683 PMCID: PMC9356071 DOI: 10.1038/s41387-022-00213-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Branched-chain amino acid (BCAA) catabolism has been considered to have an emerging role in the pathogenesis of metabolic disturbances in obesity and type 2 diabetes (T2D). Several studies showed elevated plasma BCAA levels in humans with insulin resistance and patients with T2D, although the underlying reason is unknown. Dysfunctional BCAA catabolism could theoretically be an underlying factor. In vitro and animal work collectively show that modulation of the BCAA catabolic pathway alters key metabolic processes affecting glucose homeostasis, although an integrated understanding of tissue-specific BCAA catabolism remains largely unknown, especially in humans. Proof-of-concept studies in rodents -and to a lesser extent in humans – strongly suggest that enhancing BCAA catabolism improves glucose homeostasis in metabolic disorders, such as obesity and T2D. In this review, we discuss several hypothesized mechanistic links between BCAA catabolism and insulin resistance and overview current available tools to modulate BCAA catabolism in vivo. Furthermore, this review considers whether enhancing BCAA catabolism forms a potential future treatment strategy to promote metabolic health in insulin resistance and T2D.
Collapse
Affiliation(s)
- Froukje Vanweert
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|