1
|
Whelehan G, Bello O, Hakim O, Ladwa M, Umpleby AM, Amiel SA, Bodicoat DH, Goff LM. Ethnic differences in the relationship between ectopic fat deposition and insulin sensitivity in Black African and White European men across a spectrum of glucose tolerance. Diabetes Obes Metab 2024; 26:5211-5221. [PMID: 39149769 DOI: 10.1111/dom.15867] [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: 04/29/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024]
Abstract
AIM To examine the hypothesis that there would be ethnic differences in the relationship between ectopic fat and tissue-specific insulin resistance (IR) across a spectrum of glucose tolerance in Black African (BA) and White European (WE) men. MATERIALS AND METHODS Fifty-three WE men (23/10/20 normal glucose tolerance [NGT]/impaired glucose tolerance [IGT]/type 2 diabetes [T2D]) and 48 BA men (20/10/18, respectively) underwent a two-step hyperinsulinaemic-euglycaemic clamp with infusion of D-[6,6-2H2]-glucose and [2H5]-glycerol to assess hepatic, peripheral and adipose tissue IR. Magnetic resonance imaging was used to measure subcutaneous adipose tissue, visceral adipose tissue (VAT) and intrahepatic lipid (IHL). Associations between ectopic fat and IR were assessed using linear regression models. RESULTS There were no differences in tissue-specific IR between ethnic groups at any stage of glucose tolerance. VAT level was consistently lower in the BA population; NGT (p = 0.013), IGT (p = 0.006) and T2D (p = 0.015). IHL was also lower in the BA compared with the WE men (p = 0.013). VAT and IHL levels were significantly associated with hepatic IR in the BA population (p = 0.001) and with peripheral IR in the WE population (p = 0.027). CONCLUSIONS The present study suggests that BA and WE men exhibit the same degree of IR across a glucose tolerance continuum, but with lower VAT and IHL levels in the BA population, suggesting that IR may be driven by a mechanism other than increased ectopic fat accumulation in BA men.
Collapse
Affiliation(s)
- Gráinne Whelehan
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, UK
| | - Oluwatoyosi Bello
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Olah Hakim
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Meera Ladwa
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - A Margot Umpleby
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Stephanie A Amiel
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | | - Louise M Goff
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, UK
| |
Collapse
|
2
|
Sousa D, Magalhães CC, Matafome P, Pereira SP. Adipose tissue-liver cross-talk: a route to hepatic dysfunction in pregnant women with obesity. Biosci Rep 2024; 44:BSR20231679. [PMID: 39083072 PMCID: PMC11327218 DOI: 10.1042/bsr20231679] [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: 02/20/2024] [Revised: 05/24/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Obesity during pregnancy has been escalating, becoming a huge problem that poses consequences not only for the health of the offspring but also for the maternal well-being. Women's adipose and hepatic tissue metabolism undergoes significant changes during the gestational period. During pregnancy, obesity is a primary instigator of steatosis, increasing the risk of non-alcholic fatty liver disease (NAFLD), now recognized under the updated nomenclature metabolic dysfunction-associated steatotic liver disease (MASLD). Pregnant women with obesity present higher levels of free fatty acids and glucose, reduction in insulin sensitivity, and adipose tissue endocrine dysregulation. Furthermore, obesity-induced modifications in clock genes and lipid-associated gene expression within adipose tissue disrupt crucial metabolic adaptations, potentially culminating in adipose tissue dysfunction. Thus, the liver experiences increased exposure to free fatty acids through the portal vein. Higher uptake of free fatty acids into the liver disrupts hepatic lipid oxidation while enhances lipogenesis, thereby predisposing to ectopic fat deposition within the liver. This review focuses on the obesity-induced changes during pregnancy in both liver and adipose tissue metabolism, elucidating how the metabolic crosstalk between these two organs can be dysregulated in pregnant women living with obesity.
Collapse
Affiliation(s)
- Diana Sousa
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carina C Magalhães
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Polytechnic University of Coimbra, Coimbra Health School, Rua 5 de Outubro-S. Martinho do Bispo, 3046-854 Coimbra, Portugal
| | - Susana P Pereira
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra,3004-504 Coimbra, Portugal
- CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra; 3004-517 Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory of for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
3
|
Reed JN, Huang J, Li Y, Ma L, Banka D, Wabitsch M, Wang T, Ding W, Björkegren JL, Civelek M. Systems genetics analysis of human body fat distribution genes identifies adipocyte processes. Life Sci Alliance 2024; 7:e202402603. [PMID: 38702075 PMCID: PMC11068934 DOI: 10.26508/lsa.202402603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
Excess abdominal fat is a sexually dimorphic risk factor for cardio-metabolic disease and is approximated by the waist-to-hip ratio adjusted for body mass index (WHRadjBMI). Whereas this trait is highly heritable, few causal genes are known. We aimed to identify novel drivers of WHRadjBMI using systems genetics. We used two independent cohorts of adipose tissue gene expression and constructed sex- and depot-specific Bayesian networks to model gene-gene interactions from 8,492 genes. Using key driver analysis, we identified genes that, in silico and putatively in vitro, regulate many others. 51-119 key drivers in each network were replicated in both cohorts. In other cell types, 23 of these genes are found in crucial adipocyte pathways: Wnt signaling or mitochondrial function. We overexpressed or down-regulated seven key driver genes in human subcutaneous pre-adipocytes. Key driver genes ANAPC2 and RSPO1 inhibited adipogenesis, whereas PSME3 increased adipogenesis. RSPO1 increased Wnt signaling activity. In differentiated adipocytes, MIGA1 and UBR1 down-regulation led to mitochondrial dysfunction. These five genes regulate adipocyte function, and we hypothesize that they regulate fat distribution.
Collapse
Affiliation(s)
- Jordan N Reed
- https://ror.org/0153tk833 Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- https://ror.org/0153tk833 Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jiansheng Huang
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Yong Li
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Lijiang Ma
- https://ror.org/04a9tmd77 Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dhanush Banka
- https://ror.org/0153tk833 Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Centre, Ulm, Germany
| | - Tianfang Wang
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Wen Ding
- Novo Nordisk Research Center China, Novo Nordisk A/S, Beijing, China
| | - Johan Lm Björkegren
- https://ror.org/04a9tmd77 Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Mete Civelek
- https://ror.org/0153tk833 Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- https://ror.org/0153tk833 Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
4
|
Tuifua TS, Kapoor B, Partovi S, Shah SN, Bullen JA, Enders J, Laique S, Levitin A, Gadani S. Prediction of Mortality and Hepatic Encephalopathy after Transjugular Intrahepatic Portosystemic Shunt Placement: Baseline and Longitudinal Body Composition Measurement. J Vasc Interv Radiol 2024:S1051-0443(24)00025-3. [PMID: 38244917 DOI: 10.1016/j.jvir.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 01/22/2024] Open
Abstract
PURPOSE To investigate effects of baseline and early longitudinal body composition changes on mortality and hepatic encephalopathy (HE) after transjugular intrahepatic portosystemic shunt (TIPS). MATERIALS AND METHODS This is a case-control study with analysis of a TIPS registry (1995-2020) including data from patients with cirrhosis with computed tomography (CT) scans obtained within 1 month before and 3 months after TIPS. Core muscle area (CMA), macroscopic subcutaneous adipose tissue (mSAT), macroscopic visceral adipose tissue (mVAT) area, and muscle adiposity index (MAI) on CT were obtained. Multipredictor Cox proportional hazards models were used to assess the effect of body composition variables on mortality or HE. RESULTS In total, 280 patients (158 men; median age, 57.0 years; median Model for End-stage Liver Disease-sodium [MELD-Na] score, 14.0) were included. Thirty-four patients had post-TIPS imaging. Median baseline CMA was 68.3 cm2 (interquartile range, 57.7-83.5 cm2). Patients with higher baseline CMA had decreased risks of mortality (hazard ratio [HR]: 0.82; P = .04) and HE (HR: 0.82; P = .009). It improved prediction of mortality over MELD-Na and post-TIPS right atrial pressure alone (confidence interval = 0.729). An increase in CMA (HR: 0.60; P = .043) and mSAT (HR: 0.86; P = .022) or decrease in MAI (HR: 1.50; P = .049) from before to after TIPS was associated with a decreased risk of mortality. An increase in mSAT was associated with an increased risk of HE (HR: 1.11; P = .04). CONCLUSIONS CMA on CT scan 1 month before TIPS placement predicts mortality and HE in patients with cirrhosis. Changes in body composition on CT measured 3 months after TIPS placement independently predict mortality and HE.
Collapse
Affiliation(s)
- Tisileli S Tuifua
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, EC-10 Cleveland Clinic, Cleveland, Ohio; Imaging Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | | | - Sasan Partovi
- Imaging Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Shetal N Shah
- Imaging Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Jennifer A Bullen
- Department of Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Jacob Enders
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, EC-10 Cleveland Clinic, Cleveland, Ohio; Imaging Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Sobia Laique
- Digestive Diseases and Surgery Institute, Section of Hepatology, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Abraham Levitin
- Imaging Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Sameer Gadani
- Imaging Institute, Cleveland Clinic Foundation, Cleveland, Ohio.
| |
Collapse
|
5
|
Marketou ME, Buechler NS, Fragkiadakis K, Plevritaki A, Zervakis S, Maragkoudakis S, Tsiavos A, Simantirakis E, Kochiadakis G. Visceral fat and cardiometabolic future in children and adolescents: a critical update. Pediatr Res 2023; 94:1639-1647. [PMID: 37402844 DOI: 10.1038/s41390-023-02709-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/26/2023] [Accepted: 04/03/2023] [Indexed: 07/06/2023]
Abstract
Cardiovascular disease (CVD) is a process whose pathogenetic mechanisms start very early in life. Recently, the importance of visceral adipose tissue (VAT) has been highlighted in the development of CVD. VAT does not always depend on body mass index (BMI) and has been implicated in unfavorable metabolic activity and cardiovascular adverse events. Abnormally high deposition of VAT is associated with metabolic syndrome, obesity-associated phenotype, and cardiometabolic risk factors. Although the importance of visceral fat has not been studied broadly or extensively in long-term studies in children and adolescents, it appears that it does not have the same behavior as in adults, it is related to the appearance of cardiac risk factors. In adolescents, it plays a role in the pathogenesis of CVD that occur later in adulthood. Excess body weight and adiposity may lead to the development of early myocardial and pathological coronary changes in childhood. The purpose of this review is to summarize the risk factors, the clinical significance, and the prognostic role of visceral obesity in children and adolescents. In addition, extensive reference is made to the most commonly used techniques for the evaluation of VAT in clinical settings. IMPACT: Visceral obesity, plays an important role in cardiovascular health from very early in an individual's life. Visceral adipose tissue (VAT) distribution is not entirely related to body mass index (BMI) and provides additional prognostic information. There is a need to pay more attention to the assessment of VAT in young people, to develop methods that would go beyond the measurement of only BMI in clinical practice and to identify individuals with excess visceral adiposity and perhaps to monitor its changes.
Collapse
Affiliation(s)
- Maria E Marketou
- Cardiology Department, Heraklion University General Hospital, Crete, Greece.
| | | | | | | | - Stelios Zervakis
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | | | - Alexandros Tsiavos
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | | | - George Kochiadakis
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| |
Collapse
|
6
|
Reed JN, Huang J, Li Y, Ma L, Banka D, Wabitsch M, Wang T, Ding W, Björkegren JLM, Civelek M. Systems genetics analysis of human body fat distribution genes identifies Wnt signaling and mitochondrial activity in adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.06.556534. [PMID: 37732278 PMCID: PMC10508754 DOI: 10.1101/2023.09.06.556534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Excess fat in the abdomen is a sexually dimorphic risk factor for cardio-metabolic disease. The relative storage between abdominal and lower-body subcutaneous adipose tissue depots is approximated by the waist-to-hip ratio adjusted for body mass index (WHRadjBMI). Genome-wide association studies (GWAS) identified 346 loci near 495 genes associated with WHRadjBMI. Most of these genes have unknown roles in fat distribution, but many are expressed and putatively act in adipose tissue. We aimed to identify novel sex- and depot-specific drivers of WHRadjBMI using a systems genetics approach. METHODS We used two independent cohorts of adipose tissue gene expression with 362 - 444 males and 147 - 219 females, primarily of European ancestry. We constructed sex- and depot- specific Bayesian networks to model the gene-gene interactions from 8,492 adipose tissue genes. Key driver analysis identified genes that, in silico and putatively in vitro, regulate many others, including the 495 WHRadjBMI GWAS genes. Key driver gene function was determined by perturbing their expression in human subcutaneous pre-adipocytes using lenti-virus or siRNA. RESULTS 51 - 119 key drivers in each network were replicated in both cohorts. We used single-cell expression data to select replicated key drivers expressed in adipocyte precursors and mature adipocytes, prioritized genes which have not been previously studied in adipose tissue, and used public human and mouse data to nominate 53 novel key driver genes (10 - 21 from each network) that may regulate fat distribution by altering adipocyte function. In other cell types, 23 of these genes are found in crucial adipocyte pathways: Wnt signaling or mitochondrial function. We selected seven genes whose expression is highly correlated with WHRadjBMI to further study their effects on adipogenesis/Wnt signaling (ANAPC2, PSME3, RSPO1, TYRO3) or mitochondrial function (C1QTNF3, MIGA1, PSME3, UBR1).Adipogenesis was inhibited in cells overexpressing ANAPC2 and RSPO1 compared to controls. RSPO1 results are consistent with a positive correlation between gene expression in the subcutaneous depot and WHRadjBMI, therefore lower relative storage in the subcutaneous depot. RSPO1 inhibited adipogenesis by increasing β-catenin activation and Wnt-related transcription, thus repressing PPARG and CEBPA. PSME3 overexpression led to more adipogenesis than controls. In differentiated adipocytes, MIGA1 and UBR1 downregulation led to mitochondrial dysfunction, with lower oxygen consumption than controls; MIGA1 knockdown also lowered UCP1 expression. SUMMARY ANAPC2, MIGA1, PSME3, RSPO1, and UBR1 affect adipocyte function and may drive body fat distribution.
Collapse
|
7
|
Bellavite P, Fazio S, Affuso F. A Descriptive Review of the Action Mechanisms of Berberine, Quercetin and Silymarin on Insulin Resistance/Hyperinsulinemia and Cardiovascular Prevention. Molecules 2023; 28:4491. [PMID: 37298967 PMCID: PMC10254920 DOI: 10.3390/molecules28114491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Insulin resistance (IR) and the associated hyperinsulinemia are early pathophysiological changes which, if not well treated, can lead to type 2 diabetes, endothelial dysfunction and cardiovascular disease. While diabetes care is fairly well standardized, the prevention and treatment of IR lacks a single pharmaceutical approach and many lifestyle and dietary interventions have been proposed, including a wide range of food supplements. Among the most interesting and well-known natural remedies, alkaloid berberine and the flavonol quercetin have particular relevance in the literature, while silymarin-the active principle of the Silybum marianum thistle-was traditionally used for lipid metabolism disorders and to sustain liver function. This review describes the major defects of insulin signaling leading to IR and the main properties of the three mentioned natural substances, their molecular targets and synergistic action mechanisms. The actions of berberine, quercetin and silymarin are partially superimposable as remedies against reactive oxygen intermediates generated by a high-lipid diet and by NADPH oxidase, which is triggered by phagocyte activation. Furthermore, these compounds inhibit the secretion of a battery of pro-inflammatory cytokines, modulate intestinal microbiota and are especially able to control the various disorders of the insulin receptor and post-receptor signaling systems. Although most of the evidence on the effects of berberine, quercetin and silymarin in modulating insulin resistance and preventing cardiovascular disease derive from experimental studies on animals, the amount of pre-clinical knowledge strongly suggests the need to investigate the therapeutic potential of these substances in human pathology.
Collapse
Affiliation(s)
- Paolo Bellavite
- Pathophysiology Chair, Homeopathic Medical School of Verona, 37121 Verona, Italy
| | - Serafino Fazio
- Department of Internal Medicine, University of Naples Federico II, 80138 Naples, Italy;
| | | |
Collapse
|
8
|
Mátis D, Hegyi P, Teutsch B, Tornai T, Erőss B, Pár G, Váncsa S. Improved body composition decreases the fat content in non-alcoholic fatty liver disease, a meta-analysis and systematic review of longitudinal studies. Front Med (Lausanne) 2023; 10:1114836. [PMID: 37215704 PMCID: PMC10194653 DOI: 10.3389/fmed.2023.1114836] [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: 12/02/2022] [Accepted: 04/13/2023] [Indexed: 05/24/2023] Open
Abstract
Background Based on cross-sectional studies, there is a link between body composition parameters and steatosis in non-alcoholic fatty liver disease (NAFLD). However, whether long-term changes in different body composition parameters will result in NAFLD resolution is unclear. Therefore, we aimed to summarize the literature on longitudinal studies evaluating the association between NAFLD resolution and body composition change. Methods Based on the recommendations of the Cochrane Handbook, we performed a systematic search on September 26th, 2021, in three databases: Embase, MEDLINE (via PubMed), and Cochrane Central Register of Controlled Trials (CENTRAL). Eligible studies reported on patients with NAFLD (liver fat >5%) and examined the correlation between body composition improvement and decrease in steatosis. We did not have pre-defined body composition or steatosis measurement criteria. Next, we calculated pooled correlation coefficient (r) with a 95% confidence interval (CI). Furthermore, we narratively summarized articles with other statistical methods. Results We included 15 studies in our narrative review and five in our quantitative synthesis. Based on two studies with 85 patients, we found a pooled correlation coefficient of r = 0.49 (CI: 0.22-0.69, Spearman's correlation) between the change of visceral adipose tissue and liver steatosis. Similarly, based on three studies with 175 patients, the correlation was r = 0.33 (CI: 0.19-0.46, Pearson's correlation). On the other hand, based on two studies with 163 patients, the correlation between subcutaneous adipose tissue change and liver steatosis change was r = 0.42 (CI: 0.29-0.54, Pearson's correlation). Furthermore, based on the studies in the narrative synthesis, body composition improvement was associated with steatosis resolution. Conclusions Based on the included studies, body composition improvement may be associated with a decrease in liver fat content in NAFLD. Systematic review registration Identifier: CRD42021278584.
Collapse
Affiliation(s)
- Dóra Mátis
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Brigitta Teutsch
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Tornai
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Bálint Erőss
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| | - Gabriella Pár
- Division of Gastroenterology, First Department of Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Szilárd Váncsa
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
| |
Collapse
|
9
|
Yin X, Qiu L, Long D, Lv Z, Liu Q, Wang S, Zhang W, Zhang K, Xie M. The ancient CgPEPCK-1, not CgPECK-2, evolved into a multifunctional molecule as an intracellular enzyme and extracellular PRR. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104722. [PMID: 37116769 DOI: 10.1016/j.dci.2023.104722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/11/2023]
Abstract
Phosphoenolpyruvate carboxykinase (PEPCK) is a well-known lyase involved in gluconeogenesis, while their evolution and function differentiation have not been fully understood. In this study, by constructing a phylogenetic tree to examine PEPCKs throughout the evolution from poriferans to vertebrates, Mollusk was highlighted as the only phylum to exhibit two distinct lineages, Mollusca_PEPCK-1 and Mollusca_PEPCK-2. Further study of two representative members from Crassostrea gigas (CgPEPCK-1 and CgPEPCK-2) showed that they both shared conserved sequences and structural characteristics of the catalytic enzyme, while CgPEPCK-2 displayed a higher expression level than CgPEPCK-1 in all tested tissues, and CgPEPCK-1 was specifically implicated in the immune defense against LPS stimulation and Vibrio splendidus infection. Functional analysis revealed that CgPEPCK-2 had stronger enzymatic activity than CgPEPCK-1, while CgPEPCK-1 exhibited stronger binding activity with various PAMPs, and only the protein of CgPEPCK-1 increased significantly in hemolymph during immune stimulation. All results supported that distinct sequence and function differentiations of the PEPCK gene family should have occurred since Mollusk. The more advanced evolutionary branch Mollusca_PEPCK-2 should preserve its essential function as a catalytic enzyme to be more specialized and efficient, while the ancient branch Mollusca_PEPCK-1 probably contained some members, such as CgPEPCK-1, that should be integrated into the immune system as an extracellular immune recognition receptor.
Collapse
Affiliation(s)
- Xiaoting Yin
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Limei Qiu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
| | - Dandan Long
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Zhao Lv
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Qing Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China
| | - Senyu Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; School of Marine Biology and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Weiqian Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Kexin Zhang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China; School of Marine Biology and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Mengxi Xie
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| |
Collapse
|
10
|
Mazor R, Babkin A, Littrup PJ, Alloush M, Sturek M, Byrd JP, Hernandez E, Bays H, Grunvald E, Mattar SG. Mesenteric fat cryolipolysis attenuates insulin resistance in the Ossabaw swine model of the metabolic syndrome. Surg Obes Relat Dis 2023; 19:374-383. [PMID: 36443211 PMCID: PMC10040421 DOI: 10.1016/j.soard.2022.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/26/2022] [Accepted: 10/09/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND The rising prevalence of insulin resistance (IR), metabolic syndrome, and type 2 diabetes are associated with increases in abdominal mesenteric fat. Adipocytes are sensitive to low temperatures, making cryolipolysis of mesenteric fat an attractive treatment modality to potentially reduce IR. OBJECTIVES We aimed to determine whether (1) cryolipolysis is safe in reducing the volume of the mesenteric fat and (2) reduction in mesenteric fat volume reduces indices of IR and glycemic dysfunction. SETTING Indiana University School of Medicine. METHODS A novel cooling device and method delivered cryolipolysis in a controlled manner to avoid tissue ablative temperatures. Ossabaw pigs (n = 8) were fed a high-fat diet for 9 months to develop visceral obesity, IR, and metabolic syndrome. Following laparotomy, mesenteric fat cryolipolysis (MFC) was performed in 5 pigs, while 3 served as sham surgery controls. The volume of the mesenteric fat was measured by computed tomography and compared with indices of glucose intolerance before and at 3 and 6 months postprocedure. RESULTS MFC safely reduced mesenteric fat volume by ∼30% at 3 months, which was maintained at 6 months. Body weight did not change in either the MFC or sham surgery control groups. Measure of glycemic control, insulin sensitivity, and blood pressure significantly improved after MFC compared with sham controls. CONCLUSION MFC reduces the volume of mesenteric fat and improves glycemic control in obese, IR Ossabaw pigs, without adverse effects.
Collapse
Affiliation(s)
| | | | - Peter J Littrup
- Department of Radiology, Wayne State University, Detroit, Michigan; Department of Radiology, Ascension Providence Rochester Hospital, Rochester, Michigan
| | - Mouhamad Alloush
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michael Sturek
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - James P Byrd
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Edward Hernandez
- Section of Minimally Invasive and Bariatric Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Harold Bays
- Louisville Metabolic and Atherosclerosis Research Center, Inc., Louisville, Kentucky
| | - Eduardo Grunvald
- Division of General Internal Medicine, Department of Medicine, University of California, San Diego, California; Bariatric and Metabolic Institute, Division of Minimally Invasive Surgery, Department of Surgery, University of California, San Diego, California
| | - Samer G Mattar
- Department of Surgery, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
11
|
Abstract
Insulin action is impaired in type 2 diabetes. The functions of the hormone are an integrated product of insulin secretion from pancreatic β-cells and insulin clearance by receptor-mediated endocytosis and degradation, mostly in liver (hepatocytes) and, to a lower extent, in extrahepatic peripheral tissues. Substantial evidence indicates that genetic or acquired abnormalities of insulin secretion or action predispose to type 2 diabetes. In recent years, along with the discovery of the molecular foundation of receptor-mediated insulin clearance, such as through the membrane glycoprotein CEACAM1, a consensus has begun to emerge that reduction of insulin clearance contributes to the disease process. In this review, we consider the evidence suggesting a pathogenic role for reduced insulin clearance in insulin resistance, obesity, hepatic steatosis, and type 2 diabetes.
Collapse
Affiliation(s)
- Sonia M Najjar
- Department of Biomedical Sciences and the Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA;
| | - Sonia Caprio
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology-National Research Council, Pisa, Italy
| |
Collapse
|
12
|
Streicher SA, Lim U, Park SL, Li Y, Sheng X, Hom V, Xia L, Pooler L, Shepherd J, Loo LWM, Ernst T, Buchthal S, Franke AA, Tiirikainen M, Wilkens LR, Haiman CA, Stram DO, Cheng I, Le Marchand L. Genome-wide association study of abdominal MRI-measured visceral fat: The multiethnic cohort adiposity phenotype study. PLoS One 2023; 18:e0279932. [PMID: 36607984 PMCID: PMC9821421 DOI: 10.1371/journal.pone.0279932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023] Open
Abstract
Few studies have explored the genetic underpinnings of intra-abdominal visceral fat deposition, which varies substantially by sex and race/ethnicity. Among 1,787 participants in the Multiethnic Cohort (MEC)-Adiposity Phenotype Study (MEC-APS), we conducted a genome-wide association study (GWAS) of the percent visceral adiposity tissue (VAT) area out of the overall abdominal area, averaged across L1-L5 (%VAT), measured by abdominal magnetic resonance imaging (MRI). A genome-wide significant signal was found on chromosome 2q14.3 in the sex-combined GWAS (lead variant rs79837492: Beta per effect allele = -4.76; P = 2.62 × 10-8) and in the male-only GWAS (lead variant rs2968545: (Beta = -6.50; P = 1.09 × 10-9), and one suggestive variant was found at 13q12.11 in the female-only GWAS (rs79926925: Beta = 6.95; P = 8.15 × 10-8). The negatively associated variants were most common in European Americans (T allele of rs79837492; 5%) and African Americans (C allele of rs2968545; 5%) and not observed in Japanese Americans, whereas the positively associated variant was most common in Japanese Americans (C allele of rs79926925, 5%), which was all consistent with the racial/ethnic %VAT differences. In a validation step among UK Biobank participants (N = 23,699 of mainly British and Irish ancestry) with MRI-based VAT volume, both rs79837492 (Beta = -0.026, P = 0.019) and rs2968545 (Beta = -0.028, P = 0.010) were significantly associated in men only (n = 11,524). In the MEC-APS, the association between rs79926925 and plasma sex hormone binding globulin levels reached statistical significance in females, but not in males, with adjustment for total adiposity (Beta = -0.24; P = 0.028), on the log scale. Rs79837492 and rs2968545 are located in intron 5 of CNTNAP5, and rs79926925, in an intergenic region between GJB6 and CRYL1. These novel findings differing by sex and racial/ethnic group warrant replication in additional diverse studies with direct visceral fat measurements.
Collapse
Affiliation(s)
- Samantha A. Streicher
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Unhee Lim
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - S. Lani Park
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Yuqing Li
- Department of Epidemiology and Biostatistics, University of California–San Francisco, San Francisco, California, United States of America
| | - Xin Sheng
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Victor Hom
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Lucy Xia
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Loreall Pooler
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - John Shepherd
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Lenora W. M. Loo
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Thomas Ernst
- University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Steven Buchthal
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Adrian A. Franke
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Maarit Tiirikainen
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Lynne R. Wilkens
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Daniel O. Stram
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California–San Francisco, San Francisco, California, United States of America
| | - Loïc Le Marchand
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| |
Collapse
|
13
|
Wing D, Eyler LT, Lenze EJ, Wetherell JL, Nichols JF, Meeusen R, Godino JG, Shimony JS, Snyder AZ, Nishino T, Nicol GE, Nagels G, Roelands B. Fatness, fitness and the aging brain: A cross sectional study of the associations between a physiological estimate of brain age and physical fitness, activity, sleep, and body composition. NEUROIMAGE. REPORTS 2022; 2:100146. [PMID: 36743444 PMCID: PMC9894084 DOI: 10.1016/j.ynirp.2022.100146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Introduction Changes in brain structure and function occur with aging. However, there is substantial heterogeneity both in terms of when these changes begin, and the rate at which they progress. Understanding the mechanisms and/or behaviors underlying this heterogeneity may allow us to act to target and slow negative changes associated with aging. Methods Using T1 weighted MRI images, we applied a novel algorithm to determine the physiological age of the brain (brain-predicted age) and the predicted age difference between this physiologically based estimate and chronological age (BrainPAD) to 551 sedentary adults aged 65 to 84 with self-reported cognitive complaint measured at baseline as part of a larger study. We also assessed maximal aerobic capacity with a graded exercise test, physical activity and sleep with accelerometers, and body composition with dual energy x-ray absorptiometry. Associations were explored both linearly and logistically using categorical groupings. Results Visceral Adipose Tissue (VAT), Total Sleep Time (TST) and maximal aerobic capacity all showed significant associations with BrainPAD. Greater VAT was associated with higher (i.e,. older than chronological) BrainPAD (r = 0.149 p = 0.001)Greater TST was associated with higher BrainPAD (r = 0.087 p = 0.042) and greater aerobic capacity was associated with lower BrainPAD (r = - 0.088 p = 0.040). With linear regression, both VAT and TST remained significant (p = 0.036 and 0.008 respectively). Each kg of VAT predicted a 0.741 year increase in BrainPAD, and each hour of increased TST predicted a 0.735 year increase in BrainPAD. Maximal aerobic capacity did not retain statistical significance in fully adjusted linear models. Discussion Accumulation of visceral adipose tissue and greater total sleep time, but not aerobic capacity, total daily physical activity, or sleep quantity and/or quality are associated with brains that are physiologically older than would be expected based upon chronological age alone (BrainPAD).
Collapse
Affiliation(s)
- David Wing
- Herbert Wertheim School of Public Health and Human Longevity, University of California, San Diego, United States
- Exercise and Physical Activity Resource Center (EPARC), University of California, San Diego, United States
| | - Lisa T. Eyler
- Department of Psychiatry, University of California, San Diego, United States
- San Diego Veterans Administration Health Care System, San Diego, United States
| | - Eric J. Lenze
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Julie Loebach Wetherell
- Mental Health Service, VA San Diego Healthcare System, United States
- Department of Psychiatry, University of California, San Diego, United States
| | - Jeanne F. Nichols
- Herbert Wertheim School of Public Health and Human Longevity, University of California, San Diego, United States
- Exercise and Physical Activity Resource Center (EPARC), University of California, San Diego, United States
| | - Romain Meeusen
- Human Physiology & Sports Physiotherapy Research Group, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Job G. Godino
- Herbert Wertheim School of Public Health and Human Longevity, University of California, San Diego, United States
- Exercise and Physical Activity Resource Center (EPARC), University of California, San Diego, United States
| | - Joshua S. Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Abraham Z. Snyder
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Tomoyuki Nishino
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Ginger E. Nicol
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Guy Nagels
- Department of Neurology, UZ Brussel, Brussels, Belgium
- Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Bart Roelands
- Human Physiology & Sports Physiotherapy Research Group, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
14
|
Saad RK, Ghezzawi M, Horanieh R, Khamis AM, Saunders KH, Batsis JA, Chakhtoura M. Abdominal Visceral Adipose Tissue and All-Cause Mortality: A Systematic Review. Front Endocrinol (Lausanne) 2022; 13:922931. [PMID: 36082075 PMCID: PMC9446237 DOI: 10.3389/fendo.2022.922931] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Increased abdominal visceral adipose tissue (VAT) implies an adverse cardio-metabolic profile. We examined the association of abdominal VAT parameters and all-cause mortality risk. Methods We systematically searched four databases. We performed citations/articles screening, data abstraction, and quality assessment in duplicate and independently (CRD42020205021). Results We included 12 cohorts, the majority used computed tomography to assess abdominal VAT area. Six cohorts with a mean age ≤ 65 years, examining all-cause mortality risk per increment in VAT area (cm2) or volume (cm3), showed a 11-98% relative risk increase with higher VAT parameters. However, the association lost significance after adjusting for glycemic indices, body mass index, or other fat parameters. In 4 cohorts with a mean age >65 years, the findings on mortality were inconsistent. Conversely, in two cohorts (mean age 73-77 years), a higher VAT density, was inversely proportional to VAT area, and implied a higher mortality risk. Conclusion A high abdominal VAT area seems to be associated with increased all-cause mortality in individuals ≤ 65 years, possibly mediated by metabolic complications, and not through an independent effect. This relationship is weaker and may reverse in older individuals, most likely secondary to confounding bias and reverse causality. An individual participant data meta-analysis is needed to confirm our findings, and to define an abdominal VAT area cutoff implying increased mortality risk. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=205021, identifier CRD42020205021.
Collapse
Affiliation(s)
- Randa K. Saad
- Calcium Metabolism and Osteoporosis Program, World Health Organization (WHO) Collaborating Center for Metabolic Bone Disorders, Division of Endocrinology and Metabolism, Department of Internal Medicine - American University of Beirut Medical Center, Beirut, Lebanon
- Departement of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Malak Ghezzawi
- Departement of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Renee Horanieh
- Departement of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Assem M. Khamis
- Wolfson Palliative Care Research Centre, Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Katherine H. Saunders
- Division of Endocrinology, Diabetes & Metabolism, Weill Cornell Medicine, New York, NY, United States
| | - John A. Batsis
- Division of Geriatric Medicine and Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Marlene Chakhtoura
- Calcium Metabolism and Osteoporosis Program, World Health Organization (WHO) Collaborating Center for Metabolic Bone Disorders, Division of Endocrinology and Metabolism, Department of Internal Medicine - American University of Beirut Medical Center, Beirut, Lebanon
- Departement of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
15
|
Najjar SM, Abdolahipour R, Ghadieh HE, Jahromi MS, Najjar JA, Abuamreh BAM, Zaidi S, Kumarasamy S, Muturi HT. Regulation of Insulin Clearance by Non-Esterified Fatty Acids. Biomedicines 2022; 10:biomedicines10081899. [PMID: 36009446 PMCID: PMC9405499 DOI: 10.3390/biomedicines10081899] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Insulin stores lipid in adipocytes and prevents lipolysis and the release of non-esterified fatty acids (NEFA). Excessive release of NEFA during sustained energy supply and increase in abdominal adiposity trigger systemic insulin resistance, including in the liver, a major site of insulin clearance. This causes a reduction in insulin clearance as a compensatory mechanism to insulin resistance in obesity. On the other hand, reduced insulin clearance in the liver can cause chronic hyperinsulinemia, followed by downregulation of insulin receptor and insulin resistance. Delineating the cause–effect relationship between reduced insulin clearance and insulin resistance has been complicated by the fact that insulin action and clearance are mechanistically linked to insulin binding to its receptors. This review discusses how NEFA mobilization contributes to the reciprocal relationship between insulin resistance and reduced hepatic insulin clearance, and how this may be implicated in the pathogenesis of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence: ; Tel.: +1-740-593-2376; Fax: +1-740-593-2320
| | - Raziyeh Abdolahipour
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Balamand P.O. Box 100, Lebanon
| | - Marziyeh Salehi Jahromi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John A. Najjar
- Department of Internal Medicine, College of Medicine, University of Toledo, Toledo, OH 43606, USA
| | - Basil A. M. Abuamreh
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sobia Zaidi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Sivarajan Kumarasamy
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Harrison T. Muturi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
16
|
Fetal programming in sheep: Effects on pre- and postnatal organs and glands development in lambs. Res Vet Sci 2022; 151:100-109. [PMID: 35878535 DOI: 10.1016/j.rvsc.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/14/2021] [Accepted: 07/18/2022] [Indexed: 11/20/2022]
Abstract
The present systematic review and meta-analysis aim to summarize the effects of maternal undernutrition or overnutrition during pregnancy on the absolute weight and relative weight of the organs (liver, kidneys, heart, spleen, and lung) and glands (adrenal, pancreas, and thyroid) measured during gestation, birth and the postnatal period in lambs. After completing the search, selection, and data extraction steps, the measure of effect was generated by the individual comparison of each variable response compared with the average of the control and treated group (undernutrition or overnutrition) using the DerSimonian and Laird method for random effects. The liver was the organ most affected by maternal undernutrition, as the absolute weight of the liver was reduced during pregnancy, birth, and the postnatal period. The extent of this effect is related to the duration of the intervention. Reductions in the absolute fetal weight of the lungs and spleen have also been observed. No change in organs weight were observed when the results were expressed as relative weight. For overnutrition, the fetal weight of the liver was reduced to both absolute and relative values. In contrast, the relative weight of the kidneys has been increased. For the glands analyzed, no changes in weight were observed in either scenario (absolute or relative weight). Thus, the organs are more likely to suffer weight changes, especially during pregnancy, as a result of maternal nutrition. However, this change in organ weight seems to be closely related to the reduction in body weight of the progeny as a whole.
Collapse
|
17
|
Zhang Y, He Q, Zhang W, Xiong Y, Shen S, Yang J, Ye M. Non-linear Associations Between Visceral Adiposity Index and Cardiovascular and Cerebrovascular Diseases: Results From the NHANES (1999–2018). Front Cardiovasc Med 2022; 9:908020. [PMID: 35811709 PMCID: PMC9263190 DOI: 10.3389/fcvm.2022.908020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo investigate associations between visceral adiposity index (VAI) and cardiovascular and cerebrovascular diseases (CCDs) in the American population from 1999 to 2018.MethodsData from the National Health and Nutrition Examination Survey (1998–2018) were analyzed in this study. Specifically, VAI scores were calculated using sex-specific equations that incorporate body mass index, waist circumference (WC), high-density lipoprotein (HDL), triglycerides (TG), and cholesterol. Weighted logistic regression analysis was conducted to assess the relationship between VAI tertile and increased risk of CCDs. Restricted cubic splines were used to evaluate the non-linear relationship between VAI and CCDs, such as heart failure, angina, heart attack, stroke, hypertension, and coronary heart disease. Sensitivity analysis was conducted, using VAI quartiles as independent variables.ResultsA total of 22,622 subjects aged over 20 years were included. In the fully adjusted model after controlling for covariates, the third VAI tertile was more strongly associated with CCDs than the first VAI tertile, with odds ratio (OR) and 95% confidence interval (95% CI) values for angina of 2.86, 1.68–4.85; heart attack, 1.75, 1.14–2.69; stroke, 2.01, 1.23–3.26; hypertension, 2.28, 1.86–2.78; and coronary heart disease, 1.78, 1.32–2.41; but there was no significant association with heart failure (p > 0.05). Restricted cubic splines revealed parabolic relationships between VAI score and angina (p for non-linear = 0.03), coronary heart disease (p for non-linear = 0.01), and hypertension (p for non-linear < 0.001). Sensitivity analysis indicated that the fourth VAI quartile was more strongly associated with an increased risk of angina (OR = 2.92, 95% CI, 1.49–5.69), hypertension (OR = 2.37, 95% CI, 1.90–2.97), heart attack (OR = 1.77, 95% CI, 1.09–2.88), and coronary heart disease (OR = 1.89, 95% CI, 1.24–2.86) than the first VAI quartile. VAI had superior predictive power for prevalent CCDs than other independent indicators (p < 0.05).ConclusionVisceral adiposity index score is positively correlated with angina, heart attack, stroke, hypertension, and coronary heart disease, but not heart failure, and the relationships between VAI score and angina, hypertension, and coronary heart disease are non-linear.
Collapse
Affiliation(s)
- Yangchang Zhang
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
- The Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
- Research Center for Public Health Security, Chongqing Medical University, Chongqing, China
| | - Qiang He
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Weihao Zhang
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Yang Xiong
- West China Hospital, Sichuan University, Chengdu, China
| | - Shisi Shen
- The First School of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Jialu Yang
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
- The Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
- Research Center for Public Health Security, Chongqing Medical University, Chongqing, China
| | - Mengliang Ye
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
- The Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
- Research Center for Public Health Security, Chongqing Medical University, Chongqing, China
- *Correspondence: Mengliang Ye,
| |
Collapse
|
18
|
Anti-Obesity Effect of Dyglomera® Is Associated with Activation of the AMPK Signaling Pathway in 3T3-L1 Adipocytes and Mice with High-Fat Diet-Induced Obesity. Molecules 2022; 27:molecules27103288. [PMID: 35630765 PMCID: PMC9143725 DOI: 10.3390/molecules27103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
Dyglomera® is an aqueous ethanol extract of the fruit pods of Dichrostachys glomerata, a Cameroonian spice. Several studies have shown its anti-diabetic and anti-obesity effects. However, the underlying mechanisms for such effects remain unclear. Thus, the objective of this study was to investigate the anti-obesity effect of Dyglomera® and its underlying mechanisms in mice with high-fat diet-induced obesity and 3T3-L1 adipocytes. Our results revealed that Dyglomera® inhibited adipogenesis and lipogenesis by regulating AMPK phosphorylation in white adipose tissues (WATs) and 3T3-L1 adipocytes and promoted lipolysis by increasing the expression of lipolysis-related proteins. These results suggest that Dyglomera® can be used as an effective dietary supplement for treating obesity due to its modulating effect on adipogenesis/lipogenesis and lipolysis.
Collapse
|
19
|
Non-alcoholic fatty liver disease: a multi-system disease influenced by ageing and sex, and affected by adipose tissue and intestinal function. Proc Nutr Soc 2022; 81:146-161. [DOI: 10.1017/s0029665121003815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In recent years, a wealth of factors are associated with increased risk of developing non-alcoholic fatty liver disease (NAFLD) and NAFLD is now thought to increase the risk of multiple extra-hepatic diseases. The aim of this review is first to focus on the role of ageing and sex as key, poorly understood risk factors in the development and progression of NAFLD. Secondly, we aim to discuss the roles of white adipose tissue (WAT) and intestinal dysfunction, as producers of extra-hepatic factors known to further contribute to the pathogenesis of NAFLD. Finally, we aim to summarise the role of NAFLD as a multi-system disease affecting other organ systems beyond the liver. Both increased age and male sex increase the risk of NAFLD and this may be partly driven by alterations in the distribution and function of WAT. Similarly, changes in gut microbiota composition and intestinal function with ageing and chronic overnutrition are likely to contribute to the development of NAFLD both directly (i.e. by affecting hepatic function) and indirectly via exacerbating WAT dysfunction. Consequently, the presence of NAFLD significantly increases the risk of various extra-hepatic diseases including CVD, type 2 diabetes mellitus, chronic kidney disease and certain extra-hepatic cancers. Thus changes in WAT and intestinal function with ageing and chronic overnutrition contribute to the development of NAFLD – a multi-system disease that subsequently contributes to the development of other chronic cardiometabolic diseases.
Collapse
|
20
|
McKenzie BA, Chen FL, Gruen ME, Olby NJ. Canine Geriatric Syndrome: A Framework for Advancing Research in Veterinary Geroscience. Front Vet Sci 2022; 9:853743. [PMID: 35529834 PMCID: PMC9069128 DOI: 10.3389/fvets.2022.853743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/23/2022] [Indexed: 12/26/2022] Open
Abstract
Biological aging is the single most important risk factor for disease, disability, and ultimately death in geriatric dogs. The effects of aging in companion dogs also impose significant financial and psychological burdens on their human caregivers. The underlying physiologic processes of canine aging may be occult, or early signs of aging may be ignored because of the misconception that biological aging is natural and therefore inevitable. The ability to detect, quantify, and mitigate the deleterious processes of canine aging would greatly enhance veterinary preventative medicine and animal welfare. In this paper we propose a new conceptual framework for aging in dogs, the Canine Geriatric Syndrome (CGS). CGS consists of the multiple, interrelated physical, functional, behavioral, and metabolic changes that characterize canine aging as well as the resulting clinical manifestations, including frailty, diminished quality of life, and age-associated disease. We also identify potential key components of a CGS assessment tool, a clinical instrument that would enable veterinarians to diagnose CGS and would facilitate the development and testing of interventions to prolong healthspan and lifespan in dogs by directly targeting the biological mechanisms of aging. There are many gaps in our knowledge of the mechanisms and phenotype of aging in dogs that must be bridged before a CGS assessment tool can be deployed. The conceptual framework of CGS should facilitate identifying these gaps and should stimulate research to better characterize the processes and effects of aging in dogs and to identify the most promising preventative strategies to target these.
Collapse
Affiliation(s)
| | - Frances L. Chen
- Cellular Longevity Inc., dba Loyal, San Francisco, CA, United States
| | - Margaret E. Gruen
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Natasha J. Olby
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
21
|
Lustig RH, Collier D, Kassotis C, Roepke TA, Ji Kim M, Blanc E, Barouki R, Bansal A, Cave MC, Chatterjee S, Choudhury M, Gilbertson M, Lagadic-Gossmann D, Howard S, Lind L, Tomlinson CR, Vondracek J, Heindel JJ. Obesity I: Overview and molecular and biochemical mechanisms. Biochem Pharmacol 2022; 199:115012. [PMID: 35393120 PMCID: PMC9050949 DOI: 10.1016/j.bcp.2022.115012] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a chronic, relapsing condition characterized by excess body fat. Its prevalence has increased globally since the 1970s, and the number of obese and overweight people is now greater than those underweight. Obesity is a multifactorial condition, and as such, many components contribute to its development and pathogenesis. This is the first of three companion reviews that consider obesity. This review focuses on the genetics, viruses, insulin resistance, inflammation, gut microbiome, and circadian rhythms that promote obesity, along with hormones, growth factors, and organs and tissues that control its development. It shows that the regulation of energy balance (intake vs. expenditure) relies on the interplay of a variety of hormones from adipose tissue, gastrointestinal tract, pancreas, liver, and brain. It details how integrating central neurotransmitters and peripheral metabolic signals (e.g., leptin, insulin, ghrelin, peptide YY3-36) is essential for controlling energy homeostasis and feeding behavior. It describes the distinct types of adipocytes and how fat cell development is controlled by hormones and growth factors acting via a variety of receptors, including peroxisome proliferator-activated receptor-gamma, retinoid X, insulin, estrogen, androgen, glucocorticoid, thyroid hormone, liver X, constitutive androstane, pregnane X, farnesoid, and aryl hydrocarbon receptors. Finally, it demonstrates that obesity likely has origins in utero. Understanding these biochemical drivers of adiposity and metabolic dysfunction throughout the life cycle lends plausibility and credence to the "obesogen hypothesis" (i.e., the importance of environmental chemicals that disrupt these receptors to promote adiposity or alter metabolism), elucidated more fully in the two companion reviews.
Collapse
Affiliation(s)
- Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California, San Francisco, CA 94143, United States
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Christopher Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States
| | - Troy A Roepke
- School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | - Min Ji Kim
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Etienne Blanc
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Robert Barouki
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, United States
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, United States
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland, United Kingdom
| | - Dominique Lagadic-Gossmann
- Research Institute for Environmental and Occupational Health, University of Rennes, INSERM, EHESP, Rennes, France
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Lars Lind
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States.
| |
Collapse
|
22
|
Pitere RR, van Heerden MB, Pepper MS, Ambele MA. Slc7a8 Deletion Is Protective against Diet-Induced Obesity and Attenuates Lipid Accumulation in Multiple Organs. BIOLOGY 2022; 11:311. [PMID: 35205177 PMCID: PMC8869389 DOI: 10.3390/biology11020311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 11/22/2022]
Abstract
Adipogenesis, through adipocyte hyperplasia and/or hypertrophy, leads to increased adiposity, giving rise to obesity. A genome-wide transcriptome analysis of in vitro adipogenesis in human adipose-derived stromal/stem cells identified SLC7A8 (Solute Carrier Family 7 Member 8) as a potential novel mediator. The current study has investigated the role of SLC7A8 in adipose tissue biology using a mouse model of diet-induced obesity. slc7a8 knockout (KO) and wildtype (WT) C57BL/6J mice were fed either a control diet (CD) or a high-fat diet (HFD) for 14 weeks. On the HFD, both WT and KO mice (WTHFD and KOHFD) gained significantly more weight than their CD counterparts. However, KOHFD gained significantly less weight than WTHFD. KOHFD had significantly reduced levels of glucose intolerance compared with those observed in WTHFD. KOHFD also had significantly reduced adipocyte mass and hypertrophy in inguinal, mesenteric, perigonadal, and brown adipose depots, with a corresponding decrease in macrophage infiltration. Additionally, KOHFD had decreased lipid accumulation in the liver, heart, gastrocnemius muscle, lung, and kidney. This study demonstrates that targeting slc7a8 protects against diet-induced obesity by reducing lipid accumulation in multiple organs and suggests that if targeted, has the potential to mitigate the development of obesity-associated comorbidities.
Collapse
Affiliation(s)
- Reabetswe R. Pitere
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
| | - Marlene B. van Heerden
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
| | - Melvin A. Ambele
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
23
|
Boone-Villa D, Ventura-Sobrevilla J, Aguilera-Méndez A, Jiménez-Villarreal J. The effect of adenosine monophosphate-activated protein kinase on lipolysis in adipose tissue: an historical and comprehensive review. Arch Physiol Biochem 2022; 128:7-23. [PMID: 35143739 DOI: 10.1080/13813455.2019.1661495] [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: 10/26/2022]
Abstract
CONTEXT Lipolysis is one of the most important pathways for energy management, its control in the adipose tissue (AT) is a potential therapeutic target for metabolic diseases. Adenosine Mono Phosphate-activated Protein Kinase (AMPK) is a key regulatory enzyme in lipids metabolism and a potential target for diabetes and obesity treatment. OBJECTIVE The aim of this work is to analyse the existing information on the relationship of AMPK and lipolysis in the AT. METHODS A thorough search of bibliography was performed in the databases Scopus and Web of Knowledge using the terms lipolysis, adipose tissue, and AMPK, the unrelated publications were excluded, and the documents were analysed. RESULTS Sixty-three works were found and classified in 3 categories: inhibitory effects, stimulatory effect, and diverse relationships; remarkably, the newest researches support an upregulating relationship of AMPK over lipolysis. CONCLUSION The most probable reality is that the relationship AMPK-lipolysis depends on the experimental conditions.
Collapse
Affiliation(s)
- Daniel Boone-Villa
- School of Medicine Northern Unit, Universidad Autonoma de Coahuila, Piedras Negras, México
| | | | - Asdrúbal Aguilera-Méndez
- Institute of Biological Chemistry Research, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, México
| | | |
Collapse
|
24
|
Sun J, Lv H, Li M, Zhao L, Liu Y, Zeng N, Wei X, Chen Q, Ren P, Liu Y, Zhang P, Yang Z, Zhang Z, Wang Z. How much abdominal fat do obese patients lose short term after laparoscopic sleeve gastrectomy? A quantitative study evaluated with MRI. Quant Imaging Med Surg 2021; 11:4569-4582. [PMID: 34737924 DOI: 10.21037/qims-20-1380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/28/2021] [Indexed: 11/06/2022]
Abstract
Background This study aimed to elucidate the changes in the amount of abdominal adipose tissue after laparoscopic sleeve gastrectomy in obese Chinese patients over a relatively short follow-up period and to analyze the differences in the effects of surgery between genders. Methods Ninety-one patients were enrolled in the study, including 18 males and 73 females. These patients underwent laparoscopic sleeve gastrectomy between November 2017 and November 2019. Before and short term after surgery, the areas of subcutaneous/visceral adipose tissue and the liver proton density fat fraction were calculated with upper abdominal magnetic resonance (MR) examinations. Results Approximately 100 days after surgery, the median values of weight loss and body mass index reduction were 23.1 kg and 8.1 kg/m2, respectively. The patients achieved a greater absolute loss of subcutaneous adipose tissue index than of visceral adipose tissue index (3.2×10-3 vs. 1.6×10-3, P<0.001). The amount of weight loss, body mass index loss and absolute/relative reduction in visceral adipose tissue index were much greater in males than in females (31.7 vs. 21.7 kg, P<0.001; 9.8 vs. 7.9 kg/m2, P=0.016; 2.5×10-3 vs. 1.3×10-3, P=0.007; 28.2% vs. 20.9%, P=0.029). There was a correlation between decreased amounts in subcutaneous and visceral adipose tissue in sum and weight loss (r=0.282, P=0.032). The absolute/relative reduction in visceral adipose tissue index was also correlated with absolute/relative reduction in liver proton density fat fraction (r=0.283, P=0.013; r=0.372, P=0.001). Conclusions The reductions in body weight and visceral fat were more significant in male patients. The sum of absolute reduction in subcutaneous and visceral fat deposits was correlated with weight loss, in all patients enrolled. For severely obese patients, an upper abdominal MR examination could assess the body tissue composition and how it changes after bariatric surgery.
Collapse
Affiliation(s)
- Jing Sun
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Han Lv
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mengyi Li
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Lei Zhao
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yawen Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Na Zeng
- National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xuan Wei
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qian Chen
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Pengling Ren
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yang Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Peng Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Li J, Zhu J, Tan Z, Yu Y, Luo L, Zhou W, Zhu L, Wang T, Cao T, Liu L, Bao H, Huang X, Cheng X. Visceral adiposity index is associated with arterial stiffness in hypertensive adults with normal-weight: the china H-type hypertension registry study. Nutr Metab (Lond) 2021; 18:90. [PMID: 34627312 PMCID: PMC8502385 DOI: 10.1186/s12986-021-00617-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/09/2021] [Indexed: 01/24/2023] Open
Abstract
Background Limited information is available on arterial stiffness risk among hypertensive patients with metabolically abnormal but normal weight. Visceral adiposity index (VAI) is a novel indicator for visceral fat mass and metabolism, however, whether can be used to assessed arterial stiffness in a normal-weight population remains unclear. The goal of this study was to examine the independent association of VAI with arterial stiffness in normal-weight hypertensive patients. Methods 3258 participants recruited from the China H-type Hypertension Registry Study. VAI value was calculated using sex-specific equations. High arterial stiffness was defined as baPWV ≥ 18 m/s. Multivariable regression analysis was used to identify the association of VAI with baPWV and high arterial stiffness. Results Of participants, 50.5% (1644) were males, the mean age was 65.5 (SD, 9.1) years. Mean VAI and baPWV were 2.0 (SD, 2.3) and 18.2 (SD, 3.9) m/s, respectively. For each unit increase of lg VAI in multivariable regression analysis, there was a 1.05 m/s increase in baPWV (95% CI 0.67, 1.43) and a 2.13-fold increase in the risk of high arterial stiffness (95% CI 1.59, 2.86). In all models, the VAI was consistently and significantly associated with baPWV after adjustment for different confounders. High VAI levels were stably associated with baPWV in all subgroups. Conclusions We found positive association of VAI with baPWV and high arterial stiffness in normal-weight adults with hypertension. The establishment of this association could help the arterial stiffness risk stratification in normal-weight hypertensive populations, who are frequently overlooked in preventing cardiovascular disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12986-021-00617-5.
Collapse
Affiliation(s)
- Junpei Li
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Jian Zhu
- Qiukou Health Center, Wuyuan, China
| | - Ziheng Tan
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Yun Yu
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Linfei Luo
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Wei Zhou
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.,Center for Prevention and Treatment of Cardiovascular Diseases, Nanchang University Second Affiliated Hospital, Nanchang, Jiangxi, China
| | - Linjuan Zhu
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.,Center for Prevention and Treatment of Cardiovascular Diseases, Nanchang University Second Affiliated Hospital, Nanchang, Jiangxi, China
| | - Tao Wang
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.,Center for Prevention and Treatment of Cardiovascular Diseases, Nanchang University Second Affiliated Hospital, Nanchang, Jiangxi, China
| | - Tianyu Cao
- Biological Anthropology, University of California, Santa Barbara, California, USA
| | - Lishun Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Huihui Bao
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.,Center for Prevention and Treatment of Cardiovascular Diseases, Nanchang University Second Affiliated Hospital, Nanchang, Jiangxi, China
| | - Xiao Huang
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.
| | - Xiaoshu Cheng
- Department of Cardiology, Nanchang University Second Affiliated Hospital, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China. .,Center for Prevention and Treatment of Cardiovascular Diseases, Nanchang University Second Affiliated Hospital, Nanchang, Jiangxi, China.
| |
Collapse
|
26
|
Abe T, Matsubayashi Y, Muragishi S, Yoshida A, Suganami H, Furusawa K, Fujihara K, Tanaka S, Kaku K, Sone H. Dipeptidyl peptidase-4 inhibitor, anagliptin, alters hepatic insulin clearance in relation to the glycemic status in Japanese individuals with type 2 diabetes. J Diabetes Investig 2021; 12:1805-1815. [PMID: 33751849 PMCID: PMC8504901 DOI: 10.1111/jdi.13543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS/INTRODUCTION This study investigated the impact of the dipeptidyl peptidase-4 inhibitor, anagliptin, on hepatic insulin clearance (HIC) in Japanese type 2 diabetes patients and explored its relationship to glycemic status. MATERIALS AND METHODS Data on 765 participants in anagliptin phase 2 and 3 studies were analyzed. Adjusted changes in variables during 12 weeks of anagliptin therapy were compared with a placebo. HIC was calculated as the ratio, C-peptide area under the curve 0-120 min to insulin area under the curve 0-120 min, after a meal tolerance test. To explore the effects of baseline HIC levels on variables, participants receiving anagliptin were divided according to quartiles of baseline HIC. Furthermore, multivariate analysis investigated the association between baseline HIC levels and glycemic status. RESULTS Anagliptin significantly reduced glycosylated hemoglobin levels (P < 0.001 vs placebo) and HIC levels (P < 0.01). Longer duration of diabetes, lower body mass index, higher glycosylated hemoglobin and lower insulin secretion capacity were observed with increases in baseline HIC levels. Improvements in glycosylated hemoglobin, glycoalbumin and 1,5-anhydroglucitol levels were greater in the relatively higher HIC group (baseline HIC levels ≥median) than in the lower HIC group ( CONCLUSIONS Anagliptin affected HIC levels according to HIC baseline levels. Higher baseline HIC values might result in improved hyperglycemia through reduced HIC.
Collapse
Affiliation(s)
- Takahiro Abe
- Department of Hematology, Endocrinology and MetabolismNiigata University Faculty of MedicineNiigataJapan
| | - Yasuhiro Matsubayashi
- Department of Hematology, Endocrinology and MetabolismNiigata University Faculty of MedicineNiigataJapan
| | | | - Akihiro Yoshida
- Department of Hematology, Endocrinology and MetabolismNiigata University Faculty of MedicineNiigataJapan
- Kowa Co., Ltd.TokyoJapan
| | | | | | - Kazuya Fujihara
- Department of Hematology, Endocrinology and MetabolismNiigata University Faculty of MedicineNiigataJapan
| | - Shiro Tanaka
- Department of Clinical BiostatisticsGraduate School of Medicine Kyoto UniversityKyotoJapan
| | | | - Hirohito Sone
- Department of Hematology, Endocrinology and MetabolismNiigata University Faculty of MedicineNiigataJapan
| |
Collapse
|
27
|
Takashima S, Nagamori Y, Ohata K, Oikawa T, Sugaya T, Kobatake Y, Nishii N. Clinical evaluation of urinary liver-type fatty acid-binding protein for the diagnosis of renal diseases in dogs. J Vet Med Sci 2021; 83:1465-1471. [PMID: 34526412 PMCID: PMC8498833 DOI: 10.1292/jvms.20-0698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Liver-type fatty acid-binding protein (L-FABP) is a biomarker for the early detection of renal diseases in humans. L-FABP is a cytotoxic oxidation product secreted from the proximal tubules
under ischemic and oxidative stress conditions. First, L-FABP gene expression in the kidney and liver was evaluated. Next, the urinary L-FABP concentrations in dogs with or without renal
diseases were measured using a novel enzyme-linked immunosorbent assay kit. Urinary L-FABP was normalized relative to urinary creatinine (uCre) concentrations (µg/g uCre). Finally, the
relationships between urinary L-FABP and renal biomarkers used in canine medicine or serum alanine transaminase (ALT) as an indicator of liver damage were examined. Serum and urine samples
from 94 client-owned dogs including 23 dogs with renal diseases and 71 dogs without renal diseases were used for analysis. Relative L-FABP gene expression was confirmed both in the liver and
kidney. Dogs with renal diseases had a significantly higher urinary L-FABP than those without, and its predictive cutoff value was 26 µg/g uCre. Urinary L-FABP was significantly correlated
with serum creatinine (r=0.4674, P<0.01), urea nitrogen (r=0.4907, P<0.01), urine specific gravity
(r=−0.5100, P<0.01), and urine protein/creatinine ratio (r=0.7216, P<0.01), but not with serum ALT. Hence, dogs
with a high urinary L-FABP value were more likely to have renal diseases.
Collapse
Affiliation(s)
- Satoshi Takashima
- Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Yumiko Nagamori
- Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Keiichi Ohata
- CMIC Holdings Co., Ltd., Tokyo 113-0034, Japan.,Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | | | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Yui Kobatake
- Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Naohito Nishii
- Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| |
Collapse
|
28
|
Lee SG, Kang H. Anti-Obesity and Lipid Metabolism Effects of Ulmus davidiana var. japonica in Mice Fed a High-Fat Diet. J Microbiol Biotechnol 2021; 31:1011-1021. [PMID: 34099594 PMCID: PMC9706023 DOI: 10.4014/jmb.2102.02015] [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: 02/10/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022]
Abstract
The root bark of Ulmus davidiana var. japonica (Japanese elm) is used in Korea and other East Asian countries as a traditional herbal remedy to treat a variety of inflammatory diseases and ailments such as edema, gastric cancer and mastitis. For this study, we investigated the lipid metabolism and anti-obesity efficacy of ethyl alcohol extract of Ulmus davidiana var. japonica root bark (UDE). First, HPLC was performed to quantify the level of (+)-catechin, the active ingredient of UDE. In the following experiments, cultured 3T3-L1 pre-adipocytes and high-fat diet (HFD)-fed murine model were studied for anti-obesity efficacy by testing the lipid metabolism effects of UDE and (+)-catechin. In the test using 3T3-L1 pre-adipocytes, treatment with UDE inhibited adipocyte differentiation and significantly reduced the production of adipogenic genes and transcription factors PPARγ, C/EBPα and SREBP-1c. HFD-fed, obese mice were administered with UDE (200 mg/kg per day) and (+)-catechin (30 mg/kg per day) by oral gavage for 4 weeks. Weight gain, epididymal and abdominal adipose tissue mass were significantly reduced, and a change in adipocyte size was observed in the UDE and (+)-catechin treatment groups compared to the untreated control group (***p < 0.001). Significantly lower total cholesterol and triglyceride levels were detected in UDE-treated HFD mice compared to the control, revealing the efficacy of UDE. In addition, it was found that lipid accumulation in hepatocytes was also significantly reduced after administration of UDE. These results suggest that UDE has significant anti-obesity and lipid metabolism effects through inhibition of adipocyte differentiation and adipogenesis.
Collapse
Affiliation(s)
- Sung-Gyu Lee
- Department of Medical Laboratory Science, College of Health Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Hyun Kang
- Department of Medical Laboratory Science, College of Health Science, Dankook University, Cheonan 31116, Republic of Korea,Corresponding author Phone: +82-41-550-3015 Fax: +82-41-559-7934 E-mail:
| |
Collapse
|
29
|
Shah MH, Piaggi P, Looker HC, Paddock E, Krakoff J, Chang DC. Lower insulin clearance is associated with increased risk of type 2 diabetes in Native Americans. Diabetologia 2021; 64:914-922. [PMID: 33404681 DOI: 10.1007/s00125-020-05348-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/22/2020] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Impaired insulin clearance is implicated in the pathogenesis of type 2 diabetes, but prospective evidence remains limited. Therefore, we sought to identify factors associated with the metabolic clearance rate of insulin (MCRI) and to investigate whether lower MCRI is associated with increased risk of incident type 2 diabetes. METHODS From a longitudinal cohort, 570 adult Native Americans without diabetes living in the Southwestern United States were characterised at baseline and 448 participants were monitored over a median follow-up period of 7.9 years with 146 (32%) incident cases of diabetes identified (fasting plasma glucose ≥7.0 mmol/l, 2 h plasma glucose [2-h PG] ≥11.1 mmol/l, or clinical diagnosis). At baseline, participants underwent dual-energy x-ray absorptiometry or hydrodensitometry to assess body composition, a 75 g OGTT, an IVGTT to assess acute insulin response (AIR), and a hyperinsulinaemic-euglycaemic clamp to assess MCRI and insulin action (M). RESULTS In adjusted linear models, MCRI was inversely associated with body fat percentage (r = -0.35), fasting plasma insulin (r = -0.55) and AIR (r = -0.22), and positively associated with M (r = 0.17; all p < 0.0001). In multivariable Cox proportional hazard models, lower MCRI was associated with an increased risk of diabetes after adjustment for age, sex, heritage, body fat percentage, AIR, M, fasting plasma glucose, 2-h PG, and fasting plasma insulin (HR per one-SD difference in MCRI: 0.77; 95% CI 0.61, 0.98; p = 0.03). CONCLUSIONS/INTERPRETATION Lower MCRI is associated with an unfavourable metabolic phenotype and is associated with incident type 2 diabetes independent of established risk factors. CLINICAL TRIAL REGISTRATION NUMBERS ClinicalTrials.gov NCT00339482; NCT00340132.
Collapse
Affiliation(s)
- Mujtaba H Shah
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Helen C Looker
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Ethan Paddock
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA.
| |
Collapse
|
30
|
Carpentier AC. 100 th anniversary of the discovery of insulin perspective: insulin and adipose tissue fatty acid metabolism. Am J Physiol Endocrinol Metab 2021; 320:E653-E670. [PMID: 33522398 DOI: 10.1152/ajpendo.00620.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Insulin inhibits systemic nonesterified fatty acid (NEFA) flux to a greater degree than glucose or any other metabolite. This remarkable effect is mainly due to insulin-mediated inhibition of intracellular triglyceride (TG) lipolysis in adipose tissues and is essential to prevent diabetic ketoacidosis, but also to limit the potential lipotoxic effects of NEFA in lean tissues that contribute to the development of diabetes complications. Insulin also regulates adipose tissue fatty acid esterification, glycerol and TG synthesis, lipogenesis, and possibly oxidation, contributing to the trapping of dietary fatty acids in the postprandial state. Excess NEFA flux at a given insulin level has been used to define in vivo adipose tissue insulin resistance. Adipose tissue insulin resistance defined in this fashion has been associated with several dysmetabolic features and complications of diabetes, but the mechanistic significance of this concept is not fully understood. This review focusses on the in vivo regulation of adipose tissue fatty acid metabolism by insulin and the mechanistic significance of the current definition of adipose tissue insulin resistance. One hundred years after the discovery of insulin and despite decades of investigations, much is still to be understood about the multifaceted in vivo actions of this hormone on adipose tissue fatty acid metabolism.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du CHUS, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
31
|
Iwase T, Wang X, Shrimanker TV, Kolonin MG, Ueno NT. Body composition and breast cancer risk and treatment: mechanisms and impact. Breast Cancer Res Treat 2021; 186:273-283. [PMID: 33475878 DOI: 10.1007/s10549-020-06092-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE The purpose of this review is to clarify the association of body composition with breast cancer risk and treatment, including physiological mechanisms, and to elucidate strategies for overcoming unfavorable body composition changes that relate to breast cancer progression. METHODS We have summarized updated knowledge regarding the mechanism of the negative association of altered body composition with breast cancer risk and treatment. We also review strategies for reversing unfavorable body composition based on the latest clinical trial results. RESULTS Body composition changes in patients with breast cancer typically occur during menopause or as a result of chemotherapy or endocrine therapy. Dysfunction of visceral adipose tissue (VAT) in the setting of obesity underlies insulin resistance and chronic inflammation, which can lead to breast cancer development and progression. Insulin resistance and chronic inflammation are also observed in patients with breast cancer who have sarcopenia or sarcopenic obesity. Nutritional support and a personalized exercise program are the fundamental interventions for reversing unfavorable body composition. Other interventions that have been explored in specific situations include metformin, testosterone, emerging agents that directly target the adipocyte microenvironment, and bariatric surgery. CONCLUSIONS A better understanding of the biology of body composition phenotypes is key to determining the best intervention program for patients with breast cancer.
Collapse
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Tushaar Vishal Shrimanker
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mikhail G Kolonin
- Center for Metabolic and Degenerative Diseases, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Rojas-Rodriguez R, Ziegler R, DeSouza T, Majid S, Madore AS, Amir N, Pace VA, Nachreiner D, Alfego D, Mathew J, Leung K, Moore Simas TA, Corvera S. PAPPA-mediated adipose tissue remodeling mitigates insulin resistance and protects against gestational diabetes in mice and humans. Sci Transl Med 2020; 12:eaay4145. [PMID: 33239385 PMCID: PMC8375243 DOI: 10.1126/scitranslmed.aay4145] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 04/25/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
Pregnancy is a physiological state of continuous adaptation to changing maternal and fetal nutritional needs, including a reduction of maternal insulin sensitivity allowing for appropriately enhanced glucose availability to the fetus. However, excessive insulin resistance in conjunction with insufficient insulin secretion results in gestational diabetes mellitus (GDM), greatly increasing the risk for pregnancy complications and predisposing both mothers and offspring to future metabolic disease. Here, we report a signaling pathway connecting pregnancy-associated plasma protein A (PAPPA) with adipose tissue expansion in pregnancy. Adipose tissue plays a central role in the regulation of insulin sensitivity, and we show that, in both mice and humans, pregnancy caused remodeling of adipose tissue evidenced by altered adipocyte size, vascularization, and in vitro expansion capacity. PAPPA is known to be a metalloprotease secreted by human placenta that modulates insulin-like growth factor (IGF) bioavailability through prolteolysis of IGF binding proteins (IGFBPs) 2, 4, and 5. We demonstrate that recombinant PAPPA can stimulate ex vivo human adipose tissue expansion in an IGFBP-5- and IGF-1-dependent manner. Moreover, mice lacking PAPPA displayed impaired adipose tissue remodeling, pregnancy-induced insulin resistance, and hepatic steatosis, recapitulating multiple aspects of human GDM. In a cohort of 6361 pregnant women, concentrations of circulating PAPPA are inversely correlated with glycemia and odds of developing GDM. These data identify PAPPA and the IGF signaling pathway as necessary for the regulation of maternal adipose tissue physiology and systemic glucose homeostasis, with consequences for long-term metabolic risk and potential for therapeutic use.
Collapse
Affiliation(s)
- Raziel Rojas-Rodriguez
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Rachel Ziegler
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sana Majid
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Aylin S Madore
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Nili Amir
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Veronica A Pace
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel Nachreiner
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David Alfego
- Division of Data Sciences and Technology, IT, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jomol Mathew
- Division of Data Sciences and Technology, IT, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Katherine Leung
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Tiffany A Moore Simas
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
33
|
Tanaka M, Okada H, Hashimoto Y, Kumagai M, Nishimura H, Fukui M. Intraperitoneal, but not retroperitoneal, visceral adipose tissue is associated with diabetes mellitus: a cross-sectional, retrospective pilot analysis. Diabetol Metab Syndr 2020; 12:103. [PMID: 33292449 PMCID: PMC7691054 DOI: 10.1186/s13098-020-00612-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
AIM Diabetes mellitus (DM) is associated with adverse outcomes, and visceral adipose tissue (VAT), classified into intraperitoneal VAT (IVAT) and retroperitoneal VAT (RVAT), is associated with insulin resistance. This study aimed to evaluate the association of IVAT and RVAT with the prevalence or incidence of DM. METHODS In this cross-sectional, retrospective, cohort study, the prevalence and incidence of DM was analyzed in 803 and 624 middle-aged Japanese participants, respectively. The cross-sectional area of the abdominal adipose tissue was evaluated from an unenhanced computed tomography scan at the third lumbar vertebrae, and the IVAT or RVAT was analyzed using specialized software. The areas were normalized for the square value of the participants' height in meters and described as the IVAT or RVAT area index. RESULTS The IVAT area index (adjusted odds ratio [OR], 1.04; 95% confidence intervals [CI], 1.02-1.07, per 1.0 cm2/m2) or IVAT/RVAT area ratio (1.89; 1.23-2.85, per 1.0) was independently associated with the prevalence of DM, whereas the RVAT area index was not. During a follow-up (mean) of 3.7 years, 30 participants were diagnosed with DM. The IVAT area index (adjusted hazards ratio [HR], 1.02; 95% CI 1.003-1.04, per 1.0 cm2/m2) or IVAT/RVAT area ratio (2.25; 1.40-3.43, per 1.0) was independently associated with the incidence of DM, whereas the RVAT area index was not. CONCLUSIONS IVAT, but not RVAT, is associated with the prevalence or incidence of DM.
Collapse
Affiliation(s)
- Muhei Tanaka
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Hiroshi Okada
- Department of Internal Medicine, Matsushita Memorial Hospital, Osaka, Japan
| | - Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| | - Muneaki Kumagai
- Medical Corporation Soukenkai, Nishimura Clinic, Kyoto, Japan
| | | | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566 Japan
| |
Collapse
|
34
|
Kwon EY, Choi MS. Eriocitrin Improves Adiposity and Related Metabolic Disorders in High-Fat Diet-Induced Obese Mice. J Med Food 2020; 23:233-241. [PMID: 32191577 DOI: 10.1089/jmf.2019.4638] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Eriocitrin (EC) is an abundant flavonoid in lemons, which is known as a strong antioxidant agent. This study investigated the biological and molecular mechanisms underlying the anti-obesity effect of EC in high-fat diet (HFD)-fed obese mice. C57BL/6N mice were fed an HFD (40 kcal% fat) with or without 0.005% (w/w) EC for 16 weeks. Dietary EC improved adiposity by increasing adipocyte fatty acid (FA) oxidation, energy expenditure, and mRNA expression of thermogenesis-related genes in brown adipose tissue (BAT) and skeletal muscle, whereas it also decreased lipogenesis-related gene expression in white adipose tissue. In addition to adiposity, EC prevented hepatic steatosis by diminishing lipogenesis while enhancing FA oxidation in the liver and fecal lipid excretion, which was linked to attenuation of hyperlipidemia. Moreover, EC improved insulin sensitivity by decreasing hepatic gluconeogenesis and proinflammatory responses. These findings indicate that EC may protect against diet-induced adiposity and related metabolic disorders by controlling thermogenesis of BAT and skeletal muscle, FA oxidation, lipogenesis, fecal lipid excretion, glucose utilization, and gluconeogenesis.
Collapse
Affiliation(s)
- Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, Korea.,Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu, Korea
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, Korea.,Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu, Korea
| |
Collapse
|
35
|
Dassonvalle J, Díaz-Castro F, Donoso-Barraza C, Sepúlveda C, Pino-de la Fuente F, Pino P, Espinosa A, Chiong M, Llanos M, Troncoso R. Moderate Aerobic Exercise Training Prevents the Augmented Hepatic Glucocorticoid Response Induced by High-Fat Diet in Mice. Int J Mol Sci 2020; 21:ijms21207582. [PMID: 33066464 PMCID: PMC7590042 DOI: 10.3390/ijms21207582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/22/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022] Open
Abstract
Glucocorticoids (GCs) are critical regulators of energy balance. Their deregulation is associated with the development of obesity and metabolic syndrome. However, it is not understood if obesity alters the tissue glucocorticoid receptor (GR) response, and moreover whether a moderate aerobic exercise prevents the alteration in GR response induced by obesity. Methods: To evaluate the GR response in obese mice, we fed C57BL6J mice with a high-fat diet (HFD) for 12 weeks. Before mice were sacrificed, we injected them with dexamethasone. To assess the exercise role in GR response, we fed mice an HFD and subjected them to moderate aerobic exercise three times a week. Results: We found that mice fed a high-fat diet for 12 weeks developed hepatic GC hypersensitivity without changes in the gastrocnemius or epididymal fat GR response. Therefore, moderate aerobic exercise improved glucose tolerance, increased the corticosterone plasma levels, and prevented hepatic GR hypersensitivity with an increase in epididymal fat GR response. Conclusion: Collectively, our results suggest that mice with HFD-induced obesity develop hepatic GR sensitivity, which could enhance the metabolic effects of HFD in the liver. Moreover, exercise was found to be a feasible non-pharmacological strategy to prevent the deregulation of GR response in obesity.
Collapse
Affiliation(s)
- Jonatan Dassonvalle
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnologia de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile; (J.D.); (F.D.-C.); (C.D.-B.); (C.S.); (P.P.)
| | - Francisco Díaz-Castro
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnologia de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile; (J.D.); (F.D.-C.); (C.D.-B.); (C.S.); (P.P.)
| | - Camila Donoso-Barraza
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnologia de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile; (J.D.); (F.D.-C.); (C.D.-B.); (C.S.); (P.P.)
| | - Carlos Sepúlveda
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnologia de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile; (J.D.); (F.D.-C.); (C.D.-B.); (C.S.); (P.P.)
| | - Francisco Pino-de la Fuente
- Departamento de Tecnología Medica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (F.P.-d.l.F.); (A.E.)
| | - Pamela Pino
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnologia de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile; (J.D.); (F.D.-C.); (C.D.-B.); (C.S.); (P.P.)
| | - Alejandra Espinosa
- Departamento de Tecnología Medica, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (F.P.-d.l.F.); (A.E.)
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile;
| | - Miguel Llanos
- Laboratorio de Nutrición y Regulación Metabólica, INTA, Universidad de Chile, Santiago 7830490, Chile;
| | - Rodrigo Troncoso
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnologia de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile; (J.D.); (F.D.-C.); (C.D.-B.); (C.S.); (P.P.)
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380492, Chile;
- Correspondence: ; Tel.: +56-929-781-587
| |
Collapse
|
36
|
Chiyanika C, Chan DFY, Hui SCN, So H, Deng M, Yeung DKW, Nelson EAS, Chu WCW. The relationship between pancreas steatosis and the risk of metabolic syndrome and insulin resistance in Chinese adolescents with concurrent obesity and non-alcoholic fatty liver disease. Pediatr Obes 2020; 15:e12653. [PMID: 32351030 PMCID: PMC7507143 DOI: 10.1111/ijpo.12653] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/17/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The incidence of childhood obesity and associated comorbidities are on an increasing trend worldwide. More than 340 million children and adolescents aged between 5 and 19 years old were overweight or had obesity in 2016, from which over 124 million children and adolescents (6% of girls and 8% of boys) had obesity. OBJECTIVE To describe the relationship between pancreas steatosis, body fat and the risk of metabolic syndrome, insulin resistance in Hong Kong Chinese adolescents with both obesity and non-alcoholic fatty liver disease (NAFLD). METHODS Fifty two adolescents with obesity and NAFLD were analysed (14-18 years), stratified into fatty and non-fatty pancreas groups using chemical shift encoded MRI-pancreas proton density fat fraction ≥5%. Pancreatic, abdominal subcutaneous adipose tissue (SAT)/visceral adipose tissue (VAT) volumes, biochemical and anthropometric parameters were measured. Mann-Whitney U test, multiple linear/binary logistic regression analyses and odds ratios were used. RESULTS Fifty percent had fatty pancreas, 38% had metabolic syndrome and 81% had insulin resistance. Liver proton density fat fraction (PDFF) and VAT were independent predictors of insulin resistance (P = .006, .016). Pancreas and liver PDFF were both independent predictors of beta cells dysfunction (P = .015, .050) and metabolic syndrome (P = .021, .041). Presence of fatty pancreas in obesity was associated with insulin resistance (OR = 1.58, 95% CI = 0.39-6.4) and metabolic syndrome (OR = 1.70, 95% CI = 0.53-5.5). CONCLUSION A significant causal relationship exists between fatty pancreas, fatty liver, body fat and the risk of developing metabolic syndrome and insulin resistance. KEY POINTS Fatty pancreas is a common finding in adolescents with obesity, with a prevalence rate of 50% in this study cohort. Liver PDFF and VAT are independent predictors of insulin resistance while pancreas PDFF and liver PDFF are independent predictors of both beta cells dysfunction and metabolic syndrome. Presence of fatty pancreas at imaging should not be considered as a benign finding but rather as an imaging biomarker of emerging pancreatic metabolic and endocrine dysfunction.
Collapse
Affiliation(s)
- Chileka Chiyanika
- Department of Imaging and Interventional RadiologyThe Chinese University of Hong KongHong KongChina
| | - Dorothy F. Y. Chan
- Department of PaediatricsThe Chinese University of Hong KongHong KongChina
| | - Steve C. N. Hui
- Department of Imaging and Interventional RadiologyThe Chinese University of Hong KongHong KongChina,Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Hung‐kwan So
- Department of Paediatrics and Adolescent MedicineThe University of Hong KongHong KongChina
| | - Min Deng
- Department of Imaging and Interventional RadiologyThe Chinese University of Hong KongHong KongChina
| | - David K. W. Yeung
- Department of Imaging and Interventional RadiologyThe Chinese University of Hong KongHong KongChina,Department of Clinical OncologyThe Chinese University of Hong KongHong KongChina
| | | | - Winnie C. W. Chu
- Department of Imaging and Interventional RadiologyThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
37
|
Simoni P, Guglielmi R, Aparisi Gómez MP. Imaging of body composition in children. Quant Imaging Med Surg 2020; 10:1661-1671. [PMID: 32742959 DOI: 10.21037/qims.2020.04.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Overweight and obesity in children and adolescents have become a worldwide public health concern with an ever-increasing prevalence. An excessive accumulation of intraabdominal fat tissue increases the risk of developing insulin resistance, diabetes, and cardiovascular diseases in adulthood. Body composition has a role in metabolism regulation in children and adolescents with differences between genders and age groups. Until recently, Air Displacement Plethysmography and Dual-energy X-ray Absorptiometry (DXA) have been the most common techniques used to assess body composition in children. Ultrasound (US) is an accurate, readily available, and radiation-free technique to quantify intra-abdominal fat in adults, but its use in children has not yet been validated. Computed tomography (CT) is a reliable tool to assess body composition, but its use in children should be avoided due to the significant radiation burden. Quantitative Magnetic Resonance Imaging (qMRI) provides an accurate measurement of body composition, through the quantification of the visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), and brown adipose tissue (BAT), as well as lean mass. Furthermore, qMRI provides other significant estimates such as the Proton Density Fat-Fraction of the fat tissue. This review article aims to briefly describe the state of art of the advanced imaging techniques to provide a quantitative assessment of body composition in children and adolescents.
Collapse
Affiliation(s)
- Paolo Simoni
- Pediatric Imaging, Diagnostic Imaging Department, Queen Fabiola Children's University Hospital, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Riccardo Guglielmi
- Department of Radiology, St Gallen University Hospital, Kantonal Hospital Müsterlingen, Münsterlingen, Switzerland
| | - Maria Pilar Aparisi Gómez
- Department of Radiology, Auckland City Hospital, Auckland, New Zealand.,Department of Radiology, Hospital Vithas Nueve de Octubre, Valencia, Spain
| |
Collapse
|
38
|
Bone marrow fat: friend or foe in people with diabetes mellitus? Clin Sci (Lond) 2020; 134:1031-1048. [PMID: 32337536 DOI: 10.1042/cs20200220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 12/22/2022]
Abstract
Global trends in the prevalence of overweight and obesity put the adipocyte in the focus of huge medical interest. This review highlights a new topic in adipose tissue biology, namely the emerging pathogenic role of fat accumulation in bone marrow (BM). Specifically, we summarize current knowledge about the origin and function of BM adipose tissue (BMAT), provide evidence for the association of excess BMAT with diabetes and related cardiovascular complications, and discuss potential therapeutic approaches to correct BMAT dysfunction. There is still a significant uncertainty about the origins and function of BMAT, although several subpopulations of stromal cells have been suggested to have an adipogenic propensity. BM adipocytes are higly plastic and have a distinctive capacity to secrete adipokines that exert local and endocrine functions. BM adiposity is abundant in elderly people and has therefore been interpreted as a component of the whole-body ageing process. BM senescence and BMAT accumulation has been also reported in patients and animal models with Type 2 diabetes, being more pronounced in those with ischaemic complications. Understanding the mechanisms responsible for excess and altered function of BMAT could lead to new treatments able to preserve whole-body homeostasis.
Collapse
|
39
|
Robbins JM, Herzig M, Morningstar J, Sarzynski MA, Cruz DE, Wang TJ, Gao Y, Wilson JG, Bouchard C, Rankinen T, Gerszten RE. Association of Dimethylguanidino Valeric Acid With Partial Resistance to Metabolic Health Benefits of Regular Exercise. JAMA Cardiol 2020; 4:636-643. [PMID: 31166569 DOI: 10.1001/jamacardio.2019.1573] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Importance Metabolic responses to exercise training are variable. Metabolite profiling may aid in the clinical assessment of an individual's responsiveness to exercise interventions. Objective To investigate the association between a novel circulating biomarker of hepatic fat, dimethylguanidino valeric acid (DMGV), and metabolic health traits before and after 20 weeks of endurance exercise training. Design, Setting, and Participants This study involved cross-sectional and longitudinal analyses of the Health, Risk Factors, Exercise Training, and Genetics (HERITAGE) Family Study, a 20-week, single-arm endurance exercise clinical trial performed in multiple centers between 1993 and 1997. White participants with sedentary lifestyles who were free of cardiometabolic disease were included. Metabolomic tests were performed using a liquid chromatography, tandem mass spectrometry method on plasma samples collected before and after exercise training in the HERITAGE study. Metabolomics and data analysis were performed from August 2017 to May 2018. Exposures Plasma DMGV levels. Main Outcome and Measures The association between DMGV levels and measures of body composition, plasma lipids, insulin, and glucose homeostasis before and after exercise training. Results Among the 439 participants included in analyses from HERITAGE, the mean (SD) age was 36 (15) years, 228 (51.9%) were female, and the median (interquartile range) body mass index was 25 (22-28). Baseline levels of DMGV were positively associated with body fat percentage, abdominal visceral fat, very low-density lipoprotein cholesterol, and triglycerides, and inversely associated with insulin sensitivity, low-density lipoprotein cholesterol, high-density lipoprotein size, and high-density lipoprotein cholesterol (range of β coefficients, 0.17-0.46 [SEs, 0.026-0.050]; all P < .001, after adjusting for age and sex). After adjusting for age, sex, and baseline traits, baseline DMGV levels were positively associated with changes in small high-density lipoprotein particles (β, 0.14 [95% CI, 0.05-0.23]) and inversely associated with changes in medium and total high-density lipoprotein particles (β, -0.15 [95% CI, -0.24 to -0.05] and -0.19 [95% CI, -0.28 to -0.10], respectively), apolipoprotein A1 (β, -0.14 [95% CI, -0.23 to -0.05]), and insulin sensitivity (β, -0.13; P = 3.0 × 10-3) after exercise training. Conclusions and Relevance Dimethylguanidino valeric acid is an early marker of cardiometabolic dysfunction that is associated with attenuated improvements in lipid traits and insulin sensitivity after exercise training. Levels of DMGV may identify individuals who require additional therapies beyond guideline-directed exercise to improve their metabolic health.
Collapse
Affiliation(s)
- Jeremy M Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Matthew Herzig
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Jordan Morningstar
- Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Mark A Sarzynski
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia
| | - Daniel E Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Thomas J Wang
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | - Yan Gao
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
40
|
Abstract
A peculiar category of persons with obesity lacking common metabolic disturbances has been depicted and termed as metabolically healthy obesity (MHO). Yet, although MHO patients are free of obesity-associated complications, they might not be entirely precluded from developing cardio-metabolic disorders. Among patients with morbid obesity (MO) who are referred to bariatric surgery, a subset of metabolically healthy MO (MHMO) has been identified and the question arises if these patients would benefit from surgery in terms of mitigating the peril of cardio-metabolic complications. We revisited the pathophysiological mechanisms that define MHO, the currently available data on the cardio-metabolic risk of these patients and finally we reviewed the benefits of bariatric surgery and the urge to better characterize MHMO before submission to surgery.
Collapse
Affiliation(s)
- Adriana Florinela Cătoi
- Pathophysiology Department, Faculty of Medicine, 'Iuliu Hațieganu', University of Medicine and Pharmacy Cluj-Napoca Romania, Cluj-Napoca, Romania.
| | - Luca Busetto
- Department of Medicine, University of Padova, Padua, Italy
| |
Collapse
|
41
|
Le Marchand L, Wilkens LR, Castelfranco AM, Monroe KR, Kristal BS, Cheng I, Maskarinec G, Hullar MA, Lampe JW, Shepherd JA, Franke A, Ernst T, Lim U. Circulating Biomarker Score for Visceral Fat and Risks of Incident Colorectal and Postmenopausal Breast Cancer: The Multiethnic Cohort Adiposity Phenotype Study. Cancer Epidemiol Biomarkers Prev 2020; 29:966-973. [PMID: 32132150 DOI: 10.1158/1055-9965.epi-19-1469] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/24/2019] [Accepted: 02/25/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Visceral adipose tissue (VAT) may play a greater role than subcutaneous fat in increasing cancer risk but is poorly estimated in epidemiologic studies. METHODS We developed a VAT prediction score by regression equations averaged across 100 least absolute shrinkage and selection operator models in a cross-sectional study of 1,801 older adults in the Multiethnic Cohort (MEC). The score was then used as proxy for VAT in case-control studies of postmenopausal breast (950 case-control pairs) and colorectal (831 case-control pairs) cancer in an independent sample in MEC. Abdominal MRI-derived VAT; circulating biomarkers of metabolic, hormonal, and inflammation dysfunctions; and ORs for incident cancer adjusted for BMI and other risk factors were assessed. RESULTS The final score, composed of nine biomarkers, BMI, and height, explained 11% and 15% more of the variance in VAT than BMI alone in men and women, respectively. The area under the receiver operator curve for VAT >150 cm2 was 0.90 in men and 0.86 in women. The VAT score was associated with risk of breast cancer [OR (95% confidence interval [CI]) by increasing tertiles: 1.00, 1.09 (0.86-1.39), 1.48 (1.16-1.89); P trend = 0.002] but not with colorectal cancer (P = 0.84), although an association [1.00, 0.98 (0.68-1.39), 1.24 (0.88-1.76); P trend = 0.08] was suggested for this cancer after excluding cases that occurred within 7 years of blood draw (P heterogeneity = 0.06). CONCLUSIONS The VAT score predicted risks of postmenopausal breast cancer and can be used for risk assessment in diverse populations. IMPACT These findings provide specific evidence for a role of VAT in breast cancer.
Collapse
Affiliation(s)
| | | | - Ann M Castelfranco
- Bekesy Laboratory of Neurobiology, Pacific Biosciences Research Center, University of Hawaii, Honolulu, Hawaii
| | - Kristine R Monroe
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Bruce S Kristal
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Iona Cheng
- School of Medicine, University of California-San Francisco, San Francisco, California
| | | | | | | | | | - Adrian Franke
- University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Thomas Ernst
- University of Maryland School of Medicine, Baltimore, Maryland
| | - Unhee Lim
- University of Hawaii Cancer Center, Honolulu, Hawaii.
| |
Collapse
|
42
|
Chait A, den Hartigh LJ. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front Cardiovasc Med 2020; 7:22. [PMID: 32158768 PMCID: PMC7052117 DOI: 10.3389/fcvm.2020.00022] [Citation(s) in RCA: 633] [Impact Index Per Article: 158.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue plays essential roles in maintaining lipid and glucose homeostasis. To date several types of adipose tissue have been identified, namely white, brown, and beige, that reside in various specific anatomical locations throughout the body. The cellular composition, secretome, and location of these adipose depots define their function in health and metabolic disease. In obesity, adipose tissue becomes dysfunctional, promoting a pro-inflammatory, hyperlipidemic and insulin resistant environment that contributes to type 2 diabetes mellitus (T2DM). Concurrently, similar features that result from adipose tissue dysfunction also promote cardiovascular disease (CVD) by mechanisms that can be augmented by T2DM. The mechanisms by which dysfunctional adipose tissue simultaneously promote T2DM and CVD, focusing on adipose tissue depot-specific adipokines, inflammatory profiles, and metabolism, will be the focus of this review. The impact that various T2DM and CVD treatment strategies have on adipose tissue function and body weight also will be discussed.
Collapse
Affiliation(s)
- Alan Chait
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Laura J den Hartigh
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
43
|
Barnes RA, Wong T, Ross GP, Griffiths MM, Smart CE, Collins CE, MacDonald-Wicks L, Flack JR. Excessive Weight Gain Before and During Gestational Diabetes Mellitus Management: What Is the Impact? Diabetes Care 2020; 43:74-81. [PMID: 31690637 DOI: 10.2337/dc19-0800] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 10/11/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Conventional gestational diabetes mellitus (GDM) management focuses on managing blood glucose in order to prevent adverse outcomes. We hypothesized that excessive weight gain at first presentation with GDM (excessive gestational weight gain [EGWG]) and continued EGWG (cEGWG) after commencing GDM management would increase the risk of adverse outcomes, despite treatment to optimize glycemia. RESEARCH DESIGN AND METHODS Data collected prospectively from pregnant women with GDM at a single institution were analyzed. GDM was diagnosed on the basis of Australasian Diabetes in Pregnancy Society 1998 guidelines (1992-2015). EGWG means having exceeded the upper limit of the Institute of Medicine-recommended target ranges for the entire pregnancy, by GDM presentation. The relationship between EGWG and antenatal 75-g oral glucose tolerance test (oGTT) values and adverse outcomes was evaluated. Relationships were examined between cEGWG, insulin requirements, and large-for-gestational-age (LGA) infants. RESULTS Of 3,281 pregnant women, 776 (23.6%) had EGWG. Women with EGWG had higher mean fasting plasma glucose (FPG) on oGTT (5.2 mmol/L [95% CI 5.1-5.3] vs. 5.0 mmol/L [95% CI 4.9-5.0]; P < 0.01), after adjusting for confounders, and more often received insulin therapy (47.0% vs. 33.6%; P < 0.0001), with an adjusted odds ratio (aOR) of 1.4 (95% CI 1.1-1.7; P < 0.01). aORs for each 2-kg increment of cEGWG were a 1.3-fold higher use of insulin therapy (95% CI 1.1-1.5; P < 0.001), an 8-unit increase in final daily insulin dose (95% CI 5.4-11.0; P < 0.0001), and a 1.4-fold increase in the rate of delivery of LGA infants (95% CI 1.2-1.7; P < 0.0001). CONCLUSIONS The absence of EGWG and restricting cEGWG in GDM have a mitigating effect on oGTT-based FPG, the risk of having an LGA infant, and insulin requirements.
Collapse
Affiliation(s)
- Robyn A Barnes
- Diabetes Centre, Bankstown-Lidcombe Hospital, New South Wales, Australia .,School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia
| | - Tang Wong
- Diabetes Centre, Bankstown-Lidcombe Hospital, New South Wales, Australia.,Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Glynis P Ross
- Diabetes Centre, Bankstown-Lidcombe Hospital, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | | | - Carmel E Smart
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Department of Paediatric Endocrinology and Diabetes, John Hunter Children's Hospital, Newcastle, New South Wales, Australia
| | - Clare E Collins
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre in Physical Activity and Nutrition, University of Newcastle, Callaghan, New South Wales, Australia
| | - Lesley MacDonald-Wicks
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre in Physical Activity and Nutrition, University of Newcastle, Callaghan, New South Wales, Australia
| | - Jeff R Flack
- Diabetes Centre, Bankstown-Lidcombe Hospital, New South Wales, Australia.,Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,School of Medicine, Western Sydney University, Sydney, New South Wales, Australia
| |
Collapse
|
44
|
Torretta E, Barbacini P, Al-Daghri NM, Gelfi C. Sphingolipids in Obesity and Correlated Co-Morbidities: The Contribution of Gender, Age and Environment. Int J Mol Sci 2019; 20:ijms20235901. [PMID: 31771303 PMCID: PMC6929069 DOI: 10.3390/ijms20235901] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 02/07/2023] Open
Abstract
This paper reviews our present knowledge on the contribution of ceramide (Cer), sphingomyelin (SM), dihydroceramide (DhCer) and sphingosine-1-phosphate (S1P) in obesity and related co-morbidities. Specifically, in this paper, we address the role of acyl chain composition in bodily fluids for monitoring obesity in males and females, in aging persons and in situations of environmental hypoxia adaptation. After a brief introduction on sphingolipid synthesis and compartmentalization, the node of detection methods has been critically revised as the node of the use of animal models. The latter do not recapitulate the human condition, making it difficult to compare levels of sphingolipids found in animal tissues and human bodily fluids, and thus, to find definitive conclusions. In human subjects, the search for putative biomarkers has to be performed on easily accessible material, such as serum. The serum “sphingolipidome” profile indicates that attention should be focused on specific acyl chains associated with obesity, per se, since total Cer and SM levels coupled with dyslipidemia and vitamin D deficiency can be confounding factors. Furthermore, exposure to hypoxia indicates a relationship between dyslipidemia, obesity, oxygen level and aerobic/anaerobic metabolism, thus, opening new research avenues in the role of sphingolipids.
Collapse
Affiliation(s)
- Enrica Torretta
- Department of Biomedical Sciences for Health, University of Milan, Luigi Mangiagalli 31, 20133 Milan, Italy; (E.T.); (P.B.)
| | - Pietro Barbacini
- Department of Biomedical Sciences for Health, University of Milan, Luigi Mangiagalli 31, 20133 Milan, Italy; (E.T.); (P.B.)
- Ph.D. school in Molecular and Translational Medicine, University of Milan, 20142 Milan, Italy
| | - Nasser M. Al-Daghri
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department,College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Luigi Mangiagalli 31, 20133 Milan, Italy; (E.T.); (P.B.)
- I.R.C.C.S Orthopedic Institute Galeazzi, R. Galeazzi 4, 20161 Milan, Italy
- Correspondence: ; Tel.: +39-025-033-0475
| |
Collapse
|
45
|
What Is the Role of the New Index Relative Fat Mass (RFM) in the Assessment of Nonalcoholic Fatty Liver Disease (NAFLD)? Obes Surg 2019; 30:560-568. [DOI: 10.1007/s11695-019-04213-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
46
|
Eslam M, George J. Refining the role of epicardial adipose tissue in non-alcoholic fatty liver disease. Hepatol Int 2019; 13:662-664. [PMID: 31586267 DOI: 10.1007/s12072-019-09990-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
47
|
Souvannavong-Vilivong X, Sitticharoon C, Klinjampa R, Keadkraichaiwat I, Sripong C, Chatree S, Sririwichitchai R, Lertbunnaphong T. Placental expressions and serum levels of adiponectin, visfatin, and omentin in GDM. Acta Diabetol 2019; 56:1121-1131. [PMID: 31076892 DOI: 10.1007/s00592-019-01355-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/29/2019] [Indexed: 01/16/2023]
Abstract
AIMS Adiponectin, visfatin, and omentin have been shown to be associated with insulin sensitivity and might have a role in the pathophysiology of gestational diabetes mellitus (GDM). This study aimed to (1) compare adiponectin, visfatin, and omentin mRNA expressions in placenta and their serum levels between normal pregnancy (NP) and GDM class A1 (GDMA1) pregnancy and (2) determine correlations between placental gene expressions as well as serum levels with maternal and neonatal clinical parameters in all, NP, and GDM subjects. METHODS NP subjects (n = 37), who had normal medical history during their pregnancies without diagnosis of any abnormalities and GDMA1 subjects (n = 37), who were diagnosed since they had antenatal care, were recruited when they were in labor with a gestational age of at least 34 weeks. Clinical parameters and serum adiponectin, visfatin, and omentin levels were measured in the delivery room. RESULTS GDMA1 subjects had higher serum visfatin and plasma glucose levels, but lower serum omentin levels (p < 0.05 all) compared to controls, with comparable levels of placental adiponectin, visfatin, and omentin expressions, plasma insulin, and indices of insulin sensitivity and insulin resistance. Serum visfatin was negatively correlated with neonatal weight and length in the GDM group (p < 0.05 all). Serum omentin was negatively correlated with pre-pregnancy body mass index and waist circumference only in the NP group (p < 0.05 all). Serum adiponectin was negatively correlated with maternal age and HOMA-IR in the NP group (p < 0.05 all) and with placental weight and serum omentin in the GDM group (p < 0.05 all). CONCLUSIONS In conclusion, in GDMA1, increased serum visfatin, which has insulin-mimetic effect, might be associated with a compensatory mechanism that improves the impaired insulin function. Decreased serum omentin in GDMA1, which is normally found in visceral obesity, might lead to insulin resistance and contribute to the pathophysiology of GDM.
Collapse
Affiliation(s)
- Xaynaly Souvannavong-Vilivong
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Chantacha Sitticharoon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Roongrit Klinjampa
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | - Issarawan Keadkraichaiwat
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Chanakarn Sripong
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Saimai Chatree
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Rungnapa Sririwichitchai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Tripop Lertbunnaphong
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| |
Collapse
|
48
|
New Insights into the Liver-Visceral Adipose Axis During Hepatic Resection and Liver Transplantation. Cells 2019; 8:cells8091100. [PMID: 31540413 PMCID: PMC6769706 DOI: 10.3390/cells8091100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/10/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022] Open
Abstract
In the last decade, adipose tissue has emerged as an endocrine organ with a key role in energy homeostasis. In addition, there is close crosstalk between the adipose tissue and the liver, since pro- and anti-inflammatory substances produced at the visceral adipose tissue level directly target the liver through the portal vein. During surgical procedures, including hepatic resection and liver transplantation, ischemia–reperfusion injury induces damage and regenerative failure. It has been suggested that adipose tissue is associated with both pathological or, on the contrary, with protective effects on damage and regenerative response after liver surgery. The present review aims to summarize the current knowledge on the crosstalk between the adipose tissue and the liver during liver surgery. Therapeutic strategies as well as the clinical and scientific controversies in this field are discussed. The different experimental models, such as lipectomy, to evaluate the role of adipose tissue in both steatotic and nonsteatotic livers undergoing surgery, are described. Such information may be useful for the establishment of protective strategies aimed at regulating the liver–visceral adipose tissue axis and improving the postoperative outcomes in clinical liver surgery.
Collapse
|
49
|
Tanaka T, Kishi S, Ninomiya K, Tomii D, Koseki K, Sato Y, Okuno T, Sato K, Koike H, Yahagi K, Komiyama K, Aoki J, Tanabe K. Impact of abdominal fat distribution, visceral fat, and subcutaneous fat on coronary plaque scores assessed by 320-row computed tomography coronary angiography. Atherosclerosis 2019; 287:155-161. [DOI: 10.1016/j.atherosclerosis.2019.06.910] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/11/2019] [Accepted: 06/21/2019] [Indexed: 11/28/2022]
|
50
|
Thomas DD, Corkey BE, Istfan NW, Apovian CM. Hyperinsulinemia: An Early Indicator of Metabolic Dysfunction. J Endocr Soc 2019; 3:1727-1747. [PMID: 31528832 PMCID: PMC6735759 DOI: 10.1210/js.2019-00065] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023] Open
Abstract
Hyperinsulinemia is strongly associated with type 2 diabetes. Racial and ethnic minority populations are disproportionately affected by diabetes and obesity-related complications. This mini-review provides an overview of the genetic and environmental factors associated with hyperinsulinemia with a focus on racial and ethnic differences and its metabolic consequences. The data used in this narrative review were collected through research in PubMed and reference review of relevant retrieved articles. Insulin secretion and clearance are regulated processes that influence the development and progression of hyperinsulinemia. Environmental, genetic, and dietary factors are associated with hyperinsulinemia. Certain pharmacotherapies for obesity and bariatric surgery are effective at mitigating hyperinsulinemia and are associated with improved metabolic health. Hyperinsulinemia is associated with many environmental and genetic factors that interact with a wide network of hormones. Recent studies have advanced our understanding of the factors affecting insulin secretion and clearance. Further basic and translational work on hyperinsulinemia may allow for earlier and more personalized treatments for obesity and metabolic diseases.
Collapse
Affiliation(s)
- Dylan D Thomas
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Barbara E Corkey
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Nawfal W Istfan
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Caroline M Apovian
- Department of Medicine, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| |
Collapse
|