1
|
Blüher M. Understanding Adipose Tissue Dysfunction. J Obes Metab Syndr 2024; 33:275-288. [PMID: 39734091 PMCID: PMC11704217 DOI: 10.7570/jomes24013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/08/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
Diseases affecting adipose tissue (AT) function include obesity, lipodystrophy, and lipedema, among others. Both a lack of and excess AT are associated with increased risk for developing diseases including type 2 diabetes mellitus, hypertension, obstructive sleep apnea, and some types of cancer. However, individual risk of developing cardiometabolic and other 'obesity-related' diseases is not entirely determined by fat mass. Rather than excess fat accumulation, AT dysfunction may represent the mechanistic link between obesity and comorbid diseases. There are people who remain metabolically healthy despite obesity, whereas people with normal weight or very low subcutaneous AT mass may develop typically obesity-related diseases. AT dysfunction is characterized by adipocyte hypertrophy, impaired subcutaneous AT expandability (ectopic fat deposition), hypoxia, a variety of stress, inflammatory processes, and the release of proinflammatory, diabetogenic, and atherogenic signals. Genetic and environmental factors might contribute to AT heterogeneity either alone or via interaction with intrinsic biological factors. However, many questions remain regarding the mechanisms of AT dysfunction initiation and whether and how it could be reversed. Do AT signatures define clinically relevant subtypes of obesity? Is the cellular composition of AT associated with variation in obesity phenotypes? What roles do environmental compounds play in the manifestation of AT dysfunction? Answers to these and other questions may explain AT disease mechanisms and help to define strategies for improving AT health. This review focuses on recent advances in our understanding of AT biology.
Collapse
Affiliation(s)
- Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
2
|
Liu Q, Wu X, Duan W, Pan X, Wabitsch M, Lu M, Li J, Huang LH, Zhou Z, Zhu Y. ACAT1/SOAT1 maintains adipogenic ability in preadipocytes by regulating cholesterol homeostasis. J Lipid Res 2024; 65:100680. [PMID: 39481851 PMCID: PMC11638590 DOI: 10.1016/j.jlr.2024.100680] [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: 05/15/2024] [Revised: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 11/03/2024] Open
Abstract
Maintaining cholesterol homeostasis is critical for preserving adipocyte function during the progression of obesity. Despite this, the regulatory role of cholesterol esterification in governing adipocyte expandability has been understudied. Acyl-coenzyme A (CoA):cholesterol acyltransferase/Sterol O-acyltransferase 1 (ACAT1/SOAT1) is the dominant enzyme to synthesize cholesteryl ester in most tissues. Our previous study demonstrated that knockdown of either ACAT1 or ACAT2 impaired adipogenesis. However, the underlying mechanism of how ACAT1 mediates adipogenesis remains unclear. Here, we reported that ACAT1 is the dominant isoform in white adipose tissue of both humans and mice, and knocking out ACAT1 reduced fat mass in mice. Furthermore, ACAT1-deficiency inhibited the early stage of adipogenesis via attenuating PPARγ pathway. Mechanistically, ACAT1 deficiency inhibited SREBP2-mediated cholesterol uptake and thus reduced intracellular and plasma membrane cholesterol levels during adipogenesis. Replenishing cholesterol could rescue adipogenic master gene-Pparγ's-transcription in ACAT1-deficient cells during adipogenesis. Finally, overexpression of catalytically functional ACAT1, not the catalytic-dead ACAT1, rescued cholesterol levels and efficiently rescued the transcription of PPARγ as well as the adipogenesis in ACAT1-deficient preadipocytes. In summary, our study revealed the indispensable role of ACAT1 in adipogenesis via regulating intracellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Qing Liu
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong
| | - Xiaolin Wu
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong
| | - Wei Duan
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Xiaohan Pan
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Ming Lu
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Jing Li
- Department of Computing, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong
| | - Li-Hao Huang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Liver Cancer Institute Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhangsen Zhou
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yuyan Zhu
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong; Research Institute for Future Food, The Hong Kong Polytechnic University, Kowloon, Hung Hom, Hong Kong; The Hong Kong Polytechnic University Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China.
| |
Collapse
|
3
|
LaPoint A, Singer JM, Ferguson D, Shew TM, Renkemeyer MK, Palacios HH, Field RL, Yerrathota S, Kumari R, Shankaran M, Smith GI, Yoshino J, He M, Patti GJ, Hellerstein MK, Klein S, Brestoff JR, Morris EM, Finck BN, Lutkewitte AJ. Adipocyte lipin 1 expression associates with human metabolic health and regulates systemic metabolism in mice. J Clin Invest 2024; 134:e169722. [PMID: 39405118 PMCID: PMC11601902 DOI: 10.1172/jci169722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
Dysfunctional adipose tissue is believed to promote the development of hepatic steatosis and systemic insulin resistance, but many of the mechanisms involved are still unclear. Lipin 1 catalyzes the conversion of phosphatidic acid to diacylglycerol (DAG), the penultimate step of triglyceride synthesis, which is essential for lipid storage. Herein we found that adipose tissue LPIN1 expression is decreased in people with obesity compared to lean subjects, and low LPIN1 expression correlated with multi-tissue insulin resistance and increased rates of hepatic de novo lipogenesis. Comprehensive metabolic and multi-omic phenotyping demonstrated that adipocyte-specific Lpin1-/- mice had a metabolically-unhealthy phenotype, including liver and skeletal muscle insulin resistance, hepatic steatosis, increased hepatic de novo lipogenesis, and transcriptomic signatures of metabolically associated steatohepatitis that was exacerbated by high-fat diets. We conclude that adipocyte lipin 1-mediated lipid storage is vital for preserving adipose tissue and systemic metabolic health, and its loss predisposes mice to metabolically associated steatohepatitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rachael L. Field
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sireesha Yerrathota
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Roshan Kumari
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, USA
| | | | - Jun Yoshino
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan
- Center for Integrated Kidney Research and Advance (IKRA), Shimane University, Izumo, Shimane, Japan
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gary J. Patti
- Department of Medicine and
- Department of Chemistry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, USA
| | | | - Jonathan R. Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - E. Matthew Morris
- Department of Cell Biology and Physiology and
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Children’s Healthy Lifestyles and Nutrition, Kansas City, Missouri, USA
| | | | - Andrew J. Lutkewitte
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Pharmacology, University of Kansas Medical Center, Kansas City, Kansas, USA
- KU Diabetes Institute and Kansas Center for Metabolism and Obesity, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
4
|
Lee S, Cho YK, Kim H, Choi C, Kim S, Lee YH. miR-10a regulates cell death and inflammation in adipose tissue of male mice with diet-induced obesity. Mol Metab 2024; 90:102039. [PMID: 39342992 PMCID: PMC11513492 DOI: 10.1016/j.molmet.2024.102039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
OBJECTIVE Adipose tissue remodeling plays a critical role in obesity-induced metabolic dysfunction, but the underlying molecular mechanisms remain incompletely understood. This study investigates the role of miR-10a-5p in adipose tissue inflammation and metabolic dysfunction induced by a high-fat diet (HFD). METHODS Male miR-10a knockout (KO) mice were fed a HFD to induce obesity for up to 16 weeks. RNA sequencing (RNA-seq) analysis was performed to profile mRNA expression and assess the effects of miR-10a-5p KO in gonadal white adipose tissue (gWAT). Additional analyses included immunoblotting, qPCR, histological examination, and validation of the miR-10a-5p target sequence using a dual-luciferase reporter assay. RESULTS miR-10a-5p was highly expressed in gWAT but decreased after 8 weeks of HFD feeding. Over the 16-week HFD period, miR-10a KO mice exhibited greater weight gain and reduced energy expenditure compared to wild-type (WT) controls. gWAT of miR-10a KO mice on a HFD showed an increased population of proinflammatory macrophages, elevated inflammation, and increased cell death, characterized by upregulated apoptosis and necrosis markers. This was also associated with increased triglyceride accumulation in liver. Mechanistically, the proapoptotic gene Bcl2l11 was identified as a direct target of miR-10a-5p. Loss of miR-10a-5p led to BIM-mediated adipocyte death and inflammation, contributing to mitochondrial metabolic dysregulation, increased fibrosis marker expression, and the onset of inflammation in adipose tissue. CONCLUSIONS This study demonstrates the significant role of miR-10a-5p and its downstream target BIM in regulating adipocyte death during diet-induced obesity. This signaling pathway presents a potential therapeutic target for modulating obesity-induced inflammation and cell death in adipose tissue.
Collapse
Affiliation(s)
- Sumin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Keun Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Heeseong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cheoljun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sangseob Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Zhu Q, Chen S, Funcke JB, Straub LG, Lin Q, Zhao S, Joung C, Zhang Z, Kim DS, Li N, Gliniak CM, Lee C, Cebrian-Serrano A, Pedersen L, Halberg N, Gordillo R, Kusminski CM, Scherer PE. PAQR4 regulates adipocyte function and systemic metabolic health by mediating ceramide levels. Nat Metab 2024; 6:1347-1366. [PMID: 38961186 DOI: 10.1038/s42255-024-01078-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
PAQR4 is an orphan receptor in the PAQR family with an unknown function in metabolism. Here, we identify a critical role of PAQR4 in maintaining adipose tissue function and whole-body metabolic health. We demonstrate that expression of Paqr4 specifically in adipocytes, in an inducible and reversible fashion, leads to partial lipodystrophy, hyperglycaemia and hyperinsulinaemia, which is ameliorated by wild-type adipose tissue transplants or leptin treatment. By contrast, deletion of Paqr4 in adipocytes improves healthy adipose remodelling and glucose homoeostasis in diet-induced obesity. Mechanistically, PAQR4 regulates ceramide levels by mediating the stability of ceramide synthases (CERS2 and CERS5) and, thus, their activities. Overactivation of the PQAR4-CERS axis causes ceramide accumulation and impairs adipose tissue function through suppressing adipogenesis and triggering adipocyte de-differentiation. Blocking de novo ceramide biosynthesis rescues PAQR4-induced metabolic defects. Collectively, our findings suggest a critical function of PAQR4 in regulating cellular ceramide homoeostasis and targeting PAQR4 offers an approach for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jan-Bernd Funcke
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Leon G Straub
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qian Lin
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Division of Endocrinology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Chanmin Joung
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhuzhen Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dae-Seok Kim
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Na Li
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christy M Gliniak
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charlotte Lee
- Center for Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alberto Cebrian-Serrano
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Line Pedersen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
6
|
Reese-Petersen AL, Holm Nielsen S, Bülow Sand JM, Schattenberg JM, Bugianesi E, Karsdal MA. The sclerotic component of metabolic syndrome: Fibroblast activities may be the central common denominator driving organ function loss and death. Diabetes Obes Metab 2024; 26:2554-2566. [PMID: 38699780 DOI: 10.1111/dom.15615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis is a common feature of more than 50 different diseases and the cause of more than 35% of deaths worldwide, of which liver, kidney, skin, heart and, recently, lungs are receiving the most attention. Tissue changes, resulting in loss of organ function, are both a cause and consequence of disease and outcome. Fibrosis is caused by an excess deposition of extracellular matrix proteins, which over time results in impaired organ function and organ failure, and the pathways leading to increased fibroblast activation are many. This narrative review investigated the common denominator of fibrosis, fibroblasts, and the activation of fibroblasts, in response to excess energy consumption in liver, kidney, heart, skin and lung fibrosis. Fibroblasts are the main drivers of organ function loss in lung, liver, skin, heart and kidney disease. Fibroblast activation in response to excess energy consumption results in the overproduction of a range of collagens, of which types I, III and VI seem to be the essential drivers of disease progression. Fibroblast activation may be quantified in serum, enabling profiling and selection of patients. Activation of fibroblasts results in the overproduction of collagens, which deteriorates organ function. Patient profiling of fibroblast activities in serum, quantified as collagen production, may identify an organ death trajectory, better enabling identification of the right treatment for use in different metabolic interventions. As metabolically activated patients have highly elevated risk of kidney, liver and heart failure, it is essential to identify which organ to treat first and monitor organ status to correct treatment regimes. In direct alignment with this, it is essential to identify the right patients with the right organ deterioration trajectory for enrolment in clinical studies.
Collapse
Affiliation(s)
| | | | | | - Jörn M Schattenberg
- Saarland University Medical Center, Homburg, Germany
- University of the Saarland, Saarbrücken, Germany
| | | | | |
Collapse
|
7
|
Aher AP, Mittendorfer B. Fatty acids and albumin bond to control metabolic function. J Appl Physiol (1985) 2024; 136:1496-1498. [PMID: 38634504 DOI: 10.1152/japplphysiol.00262.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/19/2024] Open
Affiliation(s)
- Aman P Aher
- Department of Medicine, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Bettina Mittendorfer
- Department of Medicine, School of Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, School of Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
8
|
Pan Z, Khatry MA, Yu ML, Choudhury A, Sebastiani G, Alqahtani SA, Eslam M. MAFLD: an ideal framework for understanding disease phenotype in individuals of normal weight. Ther Adv Endocrinol Metab 2024; 15:20420188241252543. [PMID: 38808010 PMCID: PMC11131400 DOI: 10.1177/20420188241252543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/10/2024] [Indexed: 05/30/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated fatty liver disease (MAFLD) is significant, impacting almost one-third of the global population. MAFLD constitutes a primary cause of end-stage liver disease, liver cancer and the need for liver transplantation. Moreover, it has a strong association with increased mortality rates due to various extrahepatic complications, notably cardiometabolic diseases. While MAFLD is typically correlated with obesity, not all individuals with obesity develop the disease and a significant percentage of MAFLD occurs in patients without obesity, termed lean MAFLD. The clinical features, progression and underlying physiological mechanisms of patients with lean MAFLD remain inadequately characterized. The present review aims to provide a comprehensive summary of current knowledge on lean MAFLD and offer a perspective on defining MAFLD in individuals with normal weight. Key to this process is the concept of metabolic health and flexibility, which links states of dysmetabolism to the development of lean MAFLD. This perspective offers a more nuanced understanding of MAFLD and its underlying mechanisms and highlights the importance of considering the broader metabolic context in which the disease occurs. It also bridges the knowledge gap and offers insights that can inform clinical practice.
Collapse
Affiliation(s)
- Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Maryam Al Khatry
- Department of Gastroenterology, Obaidullah Hospital, Emirates Health Services, Ministry of Health, Ras Al Khaimah, United Arab Emirates
| | - Ming-Lung Yu
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ashok Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Giada Sebastiani
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, QC, Canada
| | - Saleh A. Alqahtani
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, 176 Hawkesbury Road, Westmead 2145, NSW, Australia
| |
Collapse
|
9
|
Petersen MC, Smith GI, Palacios HH, Farabi SS, Yoshino M, Yoshino J, Cho K, Davila-Roman VG, Shankaran M, Barve RA, Yu J, Stern JH, Patterson BW, Hellerstein MK, Shulman GI, Patti GJ, Klein S. Cardiometabolic characteristics of people with metabolically healthy and unhealthy obesity. Cell Metab 2024; 36:745-761.e5. [PMID: 38569471 PMCID: PMC11025492 DOI: 10.1016/j.cmet.2024.03.002] [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: 11/09/2023] [Revised: 01/06/2024] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
There is considerable heterogeneity in the cardiometabolic abnormalities associated with obesity. We evaluated multi-organ system metabolic function in 20 adults with metabolically healthy obesity (MHO; normal fasting glucose and triglycerides, oral glucose tolerance, intrahepatic triglyceride content, and whole-body insulin sensitivity), 20 adults with metabolically unhealthy obesity (MUO; prediabetes, hepatic steatosis, and whole-body insulin resistance), and 15 adults who were metabolically healthy lean. Compared with MUO, people with MHO had (1) altered skeletal muscle biology (decreased ceramide content and increased expression of genes involved in BCAA catabolism and mitochondrial structure/function); (2) altered adipose tissue biology (decreased expression of genes involved in inflammation and extracellular matrix remodeling and increased expression of genes involved in lipogenesis); (3) lower 24-h plasma glucose, insulin, non-esterified fatty acids, and triglycerides; (4) higher plasma adiponectin and lower plasma PAI-1 concentrations; and (5) decreased oxidative stress. These findings provide a framework of potential mechanisms responsible for MHO and the metabolic heterogeneity of obesity. This study was registered at ClinicalTrials.gov (NCT02706262).
Collapse
Affiliation(s)
- Max C Petersen
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA; Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, St. Louis, MO, USA
| | - Gordon I Smith
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | - Hector H Palacios
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah S Farabi
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA; Goldfarb School of Nursing at Barnes-Jewish College, St. Louis, MO, USA
| | - Mihoko Yoshino
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA; Division of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kevin Cho
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Victor G Davila-Roman
- Cardiovascular Imaging and Clinical Research Core Laboratory, Cardiovascular Division, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Ruteja A Barve
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Jinsheng Yu
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer H Stern
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Bruce W Patterson
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
10
|
Slusher AL, Nouws J, Tokoglu F, Vash-Margita A, Matthews MD, Fitch M, Shankaran M, Hellerstein MK, Caprio S. Altered extracellular matrix dynamics is associated with insulin resistance in adolescent children with obesity. Obesity (Silver Spring) 2024; 32:593-602. [PMID: 38410080 PMCID: PMC11034857 DOI: 10.1002/oby.23974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 02/28/2024]
Abstract
OBJECTIVE The objective of this study was to examine the hypothesis that abdominal and gluteal adipocyte turnover, lipid dynamics, and fibrogenesis are dysregulated among insulin-resistant (IR) compared with insulin-sensitive (IS) adolescents with obesity. METHODS Seven IS and seven IR adolescents with obesity participated in a 3-h oral glucose tolerance test and a multi-section magnetic resonance imaging scan of the abdominal region to examine body fat distribution patterns and liver fat content. An 8-week 70% deuterated water (2 H2 O) labeling protocol examined adipocyte turnover, lipid dynamics, and fibrogenesis in vivo from biopsied abdominal and gluteal fat. RESULTS Abdominal and gluteal subcutaneous adipose tissue (SAT) turnover rates of lipid components were similar among IS and IR adolescents with obesity. However, the insoluble collagen (type I, subunit α2) isoform measured from abdominal, but not gluteal, SAT was elevated in IR compared with IS individuals. In addition, abdominal insoluble collagen Iα2 was associated with ratios of visceral-to-total (visceral adipose tissue + SAT) abdominal fat and whole-body and adipose tissue insulin signaling, and it trended toward a positive association with liver fat content. CONCLUSIONS Altered extracellular matrix dynamics, but not expandability, potentially decreases abdominal SAT lipid storage capacity, contributing to the pathophysiological pathways linking adipose tissue and whole-body IR with altered ectopic storage of lipids within the liver among IR adolescents with obesity.
Collapse
Affiliation(s)
- Aaron L. Slusher
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Jessica Nouws
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Fuyuze Tokoglu
- Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Alla Vash-Margita
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Marcy D. Matthews
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Mark Fitch
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Sonia Caprio
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
11
|
Bilson J, Oquendo CJ, Read J, Scorletti E, Afolabi PR, Lord J, Bindels LB, Targher G, Mahajan S, Baralle D, Calder PC, Byrne CD, Sethi JK. Markers of adipose tissue fibrogenesis associate with clinically significant liver fibrosis and are unchanged by synbiotic treatment in patients with NAFLD. Metabolism 2024; 151:155759. [PMID: 38101770 DOI: 10.1016/j.metabol.2023.155759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND AND AIMS Subcutaneous adipose tissue (SAT) dysfunction contributes to NAFLD pathogenesis and may be influenced by the gut microbiota. Whether transcript profiles of SAT are associated with liver fibrosis and are influenced by synbiotic treatment (that changes the gut microbiome) is unknown. We investigated: (a) whether the presence of clinically significant, ≥F2 liver fibrosis associated with adipose tissue (AT) dysfunction, differential gene expression in SAT, and/or a marker of tissue fibrosis (Composite collagen gene expression (CCGE)); and (b) whether synbiotic treatment modified markers of AT dysfunction and the SAT transcriptome. METHODS Sixty-two patients with NAFLD (60 % men) were studied before and after 12 months of treatment with synbiotic or placebo and provided SAT samples. Vibration-controlled transient elastography (VCTE)-validated thresholds were used to assess liver fibrosis. RNA-sequencing and histological analysis of SAT were performed to determine differential gene expression, CCGE and the presence of collagen fibres. Regression modelling and receiver operator characteristic curve analysis were used to test associations with, and risk prediction for, ≥F2 liver fibrosis. RESULTS Patients with ≥F2 liver fibrosis (n = 24) had altered markers of AT dysfunction and a SAT gene expression signature characterised by enrichment of inflammatory and extracellular matrix-associated genes, compared to those with CONCLUSION A differential gene expression signature in SAT associates with ≥F2 liver fibrosis is explained by a measure of systemic insulin resistance and is not changed by synbiotic treatment. SAT CCGE values are a good predictor of ≥F2 liver fibrosis in NAFLD.
Collapse
Affiliation(s)
- Josh Bilson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
| | - Carolina J Oquendo
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - James Read
- School of Chemistry, Faculty of Engineering and Physical sciences, University of Southampton, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Eleonora Scorletti
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK; Division of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul R Afolabi
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
| | - Jenny Lord
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UC Louvain, Université Catholique de Louvain, Brussels, Belgium; Welbio department, WEL Research Institute, Wavre, Belgium
| | - Giovanni Targher
- Department of Medicine, University of Verona, Italy; Metabolic Diseases Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Sumeet Mahajan
- School of Chemistry, Faculty of Engineering and Physical sciences, University of Southampton, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Diana Baralle
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Christopher D Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK.
| | - Jaswinder K Sethi
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK; National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK; Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
12
|
Engin A. Adipose Tissue Hypoxia in Obesity: Clinical Reappraisal of Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:329-356. [PMID: 39287857 DOI: 10.1007/978-3-031-63657-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases β cell death. Moreover, deletion of HIF-1α worsens β cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
13
|
Gliniak CM, Pedersen L, Scherer PE. Adipose tissue fibrosis: the unwanted houseguest invited by obesity. J Endocrinol 2023; 259:e230180. [PMID: 37855264 PMCID: PMC11648981 DOI: 10.1530/joe-23-0180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/27/2023] [Indexed: 09/28/2023]
Abstract
The prevalence of obesity is increasing exponentially across the globe. The lack of effective treatment options for long-term weight loss has magnified the enormity of this problem. Studies continue to demonstrate that adipose tissue holds a biological memory, one of the most important determinant of long-term weight maintenance. This phenomenon is consistent with the metabolically dynamic role of adipose tissue: it adapts and expands to store for excess energy and serves as an endocrine organ capable of synthesizing a number of biologically active molecules that regulate metabolic homeostasis. An important component of the plasticity of adipose tissue is the extracellular matrix, essential for structural support, mechanical stability, cell signaling and function. Chronic obesity upends a delicate balance of extracellular matrix synthesis and degradation, and the ECM accumulates in such a way that prevents the plasticity and function of the diverse cell types in adipose tissue. A series of maladaptive responses among the cells in adipose tissue leads to inflammation and fibrosis, major mechanisms that explain the link between obesity and insulin resistance, risk of type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease. Adipose tissue fibrosis persists after weight loss and further enhances adipose tissue dysfunction if weight is regained. Here, we highlight the current knowledge of the cellular events governing adipose tissue ECM remodeling during the development of obesity. Our goal is to delineate the relationship more clearly between adipose tissue ECM and metabolic disease, an important step toward better defining the pathophysiology of dysfunctional adipose tissue.
Collapse
Affiliation(s)
- Christy M Gliniak
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Line Pedersen
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
14
|
Martínez–Sánchez C, Bassegoda O, Deng H, Almodóvar X, Ibarzabal A, de Hollanda A, Martínez García de la Torre R, Blaya D, Ariño S, Jiménez-Esquivel N, Aguilar-Bravo B, Vallverdú J, Montironi C, Osorio-Conles O, Fundora Y, Sánchez Moreno FJ, Gómez-Valadés AG, Aguilar-Corominas L, Soria A, Pose E, Juanola A, Cervera M, Perez M, Hernández-Gea V, Affò S, Swanson KS, Ferrer-Fàbrega J, Balibrea JM, Sancho-Bru P, Vidal J, Ginès P, Smith AM, Graupera I, Coll M. Therapeutic targeting of adipose tissue macrophages ameliorates liver fibrosis in non-alcoholic fatty liver disease. JHEP Rep 2023; 5:100830. [PMID: 37701336 PMCID: PMC10494470 DOI: 10.1016/j.jhepr.2023.100830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/02/2023] [Accepted: 06/05/2023] [Indexed: 09/14/2023] Open
Abstract
Background & Aims : The accumulation of adipose tissue macrophages (ATMs) in obesity has been associated with hepatic injury. However, the contribution of ATMs to hepatic fibrosis in non-alcoholic fatty liver disease (NAFLD) remains to be elucidated. Herein, we investigate the relationship between ATMs and liver fibrosis in patients with patients with NAFLD and evaluate the impact of modulation of ATMs over hepatic fibrosis in an experimental non-alcoholic steatohepatitis (NASH) model. Methods Adipose tissue and liver biopsies from 42 patients with NAFLD with different fibrosis stages were collected. ATMs were characterised by immunohistochemistry and flow cytometry and the correlation between ATMs and liver fibrosis stages was assessed. Selective modulation of the ATM phenotype was achieved by i.p. administration of dextran coupled with dexamethasone in diet-induced obesity and NASH murine models. Chronic administration effects were evaluated by histology and gene expression analysis in adipose tissue and liver samples. In vitro crosstalk between human ATMs and hepatic stellate cells (HSCs) and liver spheroids was performed. Results Patients with NAFLD presented an increased accumulation of pro-inflammatory ATMs that correlated with hepatic fibrosis. Long-term modulation of ATMs significantly reduced pro-inflammatory phenotype and ameliorated adipose tissue inflammation. Moreover, ATMs modulation was associated with an improvement in steatosis and hepatic inflammation and significantly reduced fibrosis progression in an experimental NASH model. In vitro, the reduction of the pro-inflammatory phenotype of human ATMs with dextran-dexamethasone treatment reduced the secretion of inflammatory chemokines and directly attenuated the pro-fibrogenic response in HSCs and liver spheroids. Conclusions Pro-inflammatory ATMs increase in parallel with fibrosis degree in patients with NAFLD and their modulation in an experimental NASH model improves liver fibrosis, uncovering the potential of ATMs as a therapeutic target to mitigate liver fibrosis in NAFLD. Impact and implications We report that human adipose tissue pro-inflammatory macrophages correlate with hepatic fibrosis in non-alcoholic fatty liver disease (NAFLD). Furthermore, the modulation of adipose tissue macrophages (ATMs) by dextran-nanocarrier conjugated with dexamethasone shifts the pro-inflammatory phenotype of ATMs to an anti-inflammatory phenotype in an experimental murine model of non-alcoholic steatohepatitis. This shift ameliorates adipose tissue inflammation, hepatic inflammation, and fibrosis. Our results highlight the relevance of adipose tissue in NAFLD pathophysiology and unveil ATMs as a potential target for NAFLD.
Collapse
Affiliation(s)
- Celia Martínez–Sánchez
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Octavi Bassegoda
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Hongping Deng
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Xènia Almodóvar
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Ainitze Ibarzabal
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ana de Hollanda
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | - Delia Blaya
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Silvia Ariño
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Natalia Jiménez-Esquivel
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Beatriz Aguilar-Bravo
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Julia Vallverdú
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Carla Montironi
- Molecular Biology Core & Pathology Department, Hospital Clínic of Barcelona, Spain
| | - Oscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Yiliam Fundora
- Department of General and Digestive Surgery, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Alicia G. Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Laia Aguilar-Corominas
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Anna Soria
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Elisa Pose
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Adrià Juanola
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Marta Cervera
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Martina Perez
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Virginia Hernández-Gea
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Silvia Affò
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
| | - Kelly S. Swanson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Joana Ferrer-Fàbrega
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Barcelona Clínic Liver Cancer Group (BCLC), IDIBAPS, Barcelona, Spain
- Hepatic Oncology Unit, Hospital Clínic, Barcelona, Spain
- Hepatobiliopancreatic Surgery and Liver and Pancreatic Transplantation Unit, Department of Surgery, Institute Clínic of Digestive and Metabolic Diseases (ICMDiM), Hospital Clínic, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Jose Maria Balibrea
- Endocrine, Metabolic & Bariatric Surgery Unit, Germans Trias i Pujol Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Pau Sancho-Bru
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Josep Vidal
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pere Ginès
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Department of General and Digestive Surgery, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Andrew M. Smith
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Carle Illinois College of Medicine, Urbana, IL, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Isabel Graupera
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacións Biomèdiques August Pi i Sunyer (FCRB-IDIABPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clínic of Barcelona, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Mar Coll
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to the accumulation of lipid laden vacuoles in hepatocytes, occurring in the context of visceral adiposity, insulin resistance and other features of the metabolic syndrome. Its more severe form (NASH, Non-Alcoholic Steatohepatitis) is becoming the leading aetiology of end-stage liver disease and hepatocellular carcinoma, and also contributes to cardiovascular disease, diabetes and extrahepatic malignancy. Management is currently limited to lifestyle modification and optimisation of the metabolic co-morbidities, with some of the drugs used for the latter also having shown some benefit for the liver. Licensed treatment modalities are currently lacking. A particular difficulty is the notorious heterogeneity of the patient population, which is poorly understood. A spectrum of disease severity associates in a non-linear way with a spectrum of severity of underlying metabolic factors. Heterogeneity of the liver in terms of mechanisms to cope with the metabolic and inflammatory stress and in terms of repair mechanisms, and a lack of knowledge hereof, further complicate the understanding of inter-individual variability. Genetic factors act as disease modifiers and potentially allow for some risk stratification, but also only explain a minor fraction of disease heterogeneity. Response to treatment shows a large variation in treatment response, again with little understanding of what is driving the absence of response in individual patients. Management can be tailored to patient's preferences in terms of diet modification, but tailoring treatment to knowledge on disease driving mechanisms in an individual patient is still in its infancy. Recent progress in analysing liver tissue as well as non-invasive tests hold, however, promise to rapidly improve our understanding of disease heterogeneity in NAFLD and provide individualised management.
Collapse
Affiliation(s)
- Sven M Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Drie Eikenstraat 655, B-2650, Edegem, Belgium.
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610, Wilrijk, Belgium.
| |
Collapse
|
16
|
Abstract
Adipose tissue exhibits a remarkable capacity to expand, contract, and remodel in response to changes in physiological and environmental conditions. Here, we describe recent advances in our understanding of how functionally distinct tissue-resident mesenchymal stromal cell subpopulations orchestrate several aspects of physiological and pathophysiological adipose tissue remodeling, with a particular focus on the adaptations that occur in response to changes in energy surplus and environmental temperature. The study of adipose tissue remodeling provides a vehicle to understand the functional diversity of stromal cells and offers a lens through which several generalizable aspects of tissue reorganization can be readily observed.
Collapse
Affiliation(s)
- Jessica Cannavino
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| | - Rana K Gupta
- Department of Medicine, Division of Endocrinology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina 27701, USA
| |
Collapse
|
17
|
Alenezi SA, Khan R, Snell L, Aboeldalyl S, Amer S. The Role of NLRP3 Inflammasome in Obesity and PCOS-A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:10976. [PMID: 37446154 DOI: 10.3390/ijms241310976] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Inflammasomes have recently been implicated in the pathogenesis of several chronic inflammatory disorders, such as diabetes and obesity. The aim of this meta-analysis was to investigate the possible role of the NLRP3 inflammasome in obesity and polycystic ovarian syndrome (PCOS). A comprehensive search of electronic databases was conducted to identify studies investigating NLRP3 its related components (Caspase 1, ASC and IL-1β) in adipose tissue and/or blood from obese individuals compared to non-obese controls. Another search was conducted for studies investigating NLRP3 in PCOS women and animal models. The ssearched databases included Medline, EMBASE, Cochrane Library, PubMed, Clinicaltrials.gov, the EU Clinical Trials Register and the WHO International Clinical Trials Register. The quality and risk of bias for the included articles were assessed using the modified Newcastle-Ottawa scale. Data were extracted and pooled using RevMan software for the calculation of the standardized mean difference (SMD) and 95% confidence interval (CI). Twelve eligible studies were included in the obesity systematic review and nine in the PCOS review. Of the obesity studies, nine (n = 270) were included in the meta-analysis, which showed a significantly higher adipose tissue NLRP3 gene expression in obese (n = 186) versus non-obese (n = 84) participants (SMD 1.07; 95% CI, 0.27, 1.87). Pooled analysis of adipose tissue IL-1β data from four studies showed significantly higher IL-1β gene expression levels in adipose tissue from 88 obese participants versus 39 non-obese controls (SMD 0.56; 95% CI, 0.13, 0.99). Meta-analysis of adipose tissue ASC data from four studies showed a significantly higher level in obese (n = 109) versus non-obese (n = 42) individuals (SMD 0.91, 95% CI, 0.30, 1.52). Of the nine PCOS articles, three were human (n = 185) and six were animal studies utilizing PCOS rat/mouse models. All studies apart from one article consistently showed upregulated NLRP3 and its components in PCOS women and animal models. In conclusion, obesity and PCOS seem to be associated with upregulated expression of NLRP3 inflammasome components. Further research is required to validate these findings and to elucidate the role of NLRP3 in obesity and PCOS.
Collapse
Affiliation(s)
- Salih Atalah Alenezi
- Division of Translational Medical Sciences, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
- Prince Mohammed Bin Abdulaziz Medical City, Ministry of Health, Riyadh 14214, Saudi Arabia
| | - Raheela Khan
- Division of Translational Medical Sciences, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| | - Lindsay Snell
- University Hospitals of Derby and Burton NHS Foundation Trust, Library & Knowledge Service, Derby DE22 3DT, UK
| | - Shaimaa Aboeldalyl
- University Hospitals of Derby and Burton NHS Foundation Trust, Obstetrics and Gynaecology, Derby DE22 3DT, UK
| | - Saad Amer
- Division of Translational Medical Sciences, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| |
Collapse
|
18
|
Lempesis IG, Georgakopoulou VE. Physiopathological mechanisms related to inflammation in obesity and type 2 diabetes mellitus. World J Exp Med 2023; 13:7-16. [PMID: 37396883 PMCID: PMC10308320 DOI: 10.5493/wjem.v13.i3.7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/15/2023] [Accepted: 04/10/2023] [Indexed: 06/16/2023] Open
Abstract
Overweight, obesity, and type 2 diabetes mellitus pose global health problems that are ever-increasing. A chronic low-grade inflammatory status and the presence of various pro-inflammatory markers either in circulation or within dysfunctional metabolic tissues are well established. The presence of these factors can, to some extent, predict disease development and progression. A central role is played by the presence of dysfunctional adipose tissue, liver dysfunction, and skeletal muscle dysfunction, which collectively contribute to the increased circulatory levels of proinflammatory factors. Weight loss and classical metabolic interventions achieve a decrease in many of these factors' circulating levels, implying that a better understanding of the processes or even the modulation of inflammation may alleviate these diseases. This review suggests that inflammation plays a significant role in the development and progression of these conditions and that measuring inflammatory markers may be useful for assessing disease risk and development of future treatment methods.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Department of Infectious Diseases-COVID-19 Unit, Laiko General Hospital, Athens 11527, Greece
| | | |
Collapse
|
19
|
Madsen S, Nelson ME, Deshpande V, Humphrey SJ, Cooke KC, Howell A, Diaz-Vegas A, Burchfield JG, Stöckli J, James DE. Deep Proteome Profiling of White Adipose Tissue Reveals Marked Conservation and Distinct Features Between Different Anatomical Depots. Mol Cell Proteomics 2023; 22:100508. [PMID: 36787876 PMCID: PMC10014311 DOI: 10.1016/j.mcpro.2023.100508] [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: 08/26/2022] [Revised: 01/26/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
White adipose tissue is deposited mainly as subcutaneous adipose tissue (SAT), often associated with metabolic protection, and abdominal/visceral adipose tissue, which contributes to metabolic disease. To investigate the molecular underpinnings of these differences, we conducted comprehensive proteomics profiling of whole tissue and isolated adipocytes from these two depots across two diets from C57Bl/6J mice. The adipocyte proteomes from lean mice were highly conserved between depots, with the major depot-specific differences encoded by just 3% of the proteome. Adipocytes from SAT (SAdi) were enriched in pathways related to mitochondrial complex I and beiging, whereas visceral adipocytes (VAdi) were enriched in structural proteins and positive regulators of mTOR presumably to promote nutrient storage and cellular expansion. This indicates that SAdi are geared toward higher catabolic activity, while VAdi are more suited for lipid storage. By comparing adipocytes from mice fed chow or Western diet (WD), we define a core adaptive proteomics signature consisting of increased extracellular matrix proteins and decreased fatty acid metabolism and mitochondrial Coenzyme Q biosynthesis. Relative to SAdi, VAdi displayed greater changes with WD including a pronounced decrease in mitochondrial proteins concomitant with upregulation of apoptotic signaling and decreased mitophagy, indicating pervasive mitochondrial stress. Furthermore, WD caused a reduction in lipid handling and glucose uptake pathways particularly in VAdi, consistent with adipocyte de-differentiation. By overlaying the proteomics changes with diet in whole adipose tissue and isolated adipocytes, we uncovered concordance between adipocytes and tissue only in the visceral adipose tissue, indicating a unique tissue-specific adaptation to sustained WD in SAT. Finally, an in-depth comparison of isolated adipocytes and 3T3-L1 proteomes revealed a high degree of overlap, supporting the utility of the 3T3-L1 adipocyte model. These deep proteomes provide an invaluable resource highlighting differences between white adipose depots that may fine-tune their unique functions and adaptation to an obesogenic environment.
Collapse
Affiliation(s)
- Søren Madsen
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Marin E Nelson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Vinita Deshpande
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Kristen C Cooke
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Anna Howell
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Alexis Diaz-Vegas
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - James G Burchfield
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Jacqueline Stöckli
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
20
|
Lempesis IG, Karlafti E, Papalexis P, Fotakopoulos G, Tarantinos K, Lekakis V, Papadakos SP, Cholongitas E, Georgakopoulou VE. COVID-19 and liver injury in individuals with obesity. World J Gastroenterol 2023; 29:908-916. [PMID: 36844135 PMCID: PMC9950870 DOI: 10.3748/wjg.v29.i6.908] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 02/10/2023] Open
Abstract
Coronavirus disease 2019 is an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 that manifests as a variety of clinical manifestations, including liver damage commonly detected by a hepatocellular pattern from liver function tests. Liver injury is associated with a worse prognosis overall. Conditions associated with the severity of the disease include obesity and cardiometabolic comorbidities, which are also associated with nonalcoholic fatty liver disease (NAFLD). The presence of NAFLD, similarly to obesity, is associated with an unfavourable impact on the coronavirus disease 2019 outcome. Individuals with these conditions could present with liver damage and elevated liver function tests due to direct viral cytotoxicity, systemic inflammation, ischemic or hypoxic liver damage or drug side effects. However, liver damage in the setting of NAFLD could also be attributed to a pre-existing chronic low-grade inflammation associated with surplus and dysfunctional adipose tissue in these individuals. Here we investigate the hypothesis that a pre-existing inflammatory status is exacerbated after severe acute respiratory syndrome coronavirus 2 infection, which embodies a second hit to the underestimated liver damage.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, United Kingdom
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht 616 6200, Netherlands
| | - Eleni Karlafti
- Department of Emergency, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki 546 21, Greece
| | - Petros Papalexis
- Unit of Endocrinology, First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
- Department of Biomedical Sciences, University of West Attica, Athens 12243, Greece
| | - George Fotakopoulos
- Department of Neurosurgery, General University Hospital of Larisa, Larisa 41221, Greece
| | | | - Vasileios Lekakis
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Stavros P Papadakos
- Department of Gastroenterology, Laiko General Hospital, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | | |
Collapse
|
21
|
Lee E, Korf H, Vidal-Puig A. An adipocentric perspective on the development and progression of non-alcoholic fatty liver disease. J Hepatol 2023; 78:1048-1062. [PMID: 36740049 DOI: 10.1016/j.jhep.2023.01.024] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
Alongside the liver, white adipose tissue (WAT) is critical in regulating systemic energy homeostasis. Although each organ has its specialised functions, they must work coordinately to regulate whole-body metabolism. Adipose tissues and the liver are relatively resilient and can adapt to an energy surplus by facilitating triglyceride (TG) storage up to a certain threshold level without significant metabolic disturbances. However, lipid storage in WAT beyond a "personalised" adiposity threshold becomes dysfunctional, leading to metabolic inflexibility, progressive inflammation, and aberrant adipokine secretion. Moreover, the failure of adipose tissue to store and mobilise lipids results in systemic knock-on lipid overload, particularly in the liver. Factors contributing to hepatic lipid overload include lipids released from WAT, dietary fat intake, and enhanced de novo lipogenesis. In contrast, extrahepatic mechanisms counteracting toxic hepatic lipid overload entail coordinated compensation through oxidation of surplus fatty acids in brown adipose tissue and storage of fatty acids as TGs in WAT. Failure of these integrated homeostatic mechanisms leads to quantitative increases and qualitative alterations to the lipidome of the liver. Initially, hepatocytes preferentially accumulate TG species leading to a relatively "benign" non-alcoholic fatty liver. However, with time, inflammatory responses ensue, progressing into more severe conditions such as non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma, in some individuals (often without an early prognostic clue). Herein, we highlight the pathogenic importance of obesity-induced "adipose tissue failure", resulting in decreased adipose tissue functionality (i.e. fat storage capacity and metabolic flexibility), in the development and progression of NAFL/NASH.
Collapse
Affiliation(s)
- Eunyoung Lee
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Department of Medical Physiology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Hannelie Korf
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium.
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK; Centro de Innvestigacion Principe Felipe, Valencia, Spain; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, China.
| |
Collapse
|
22
|
Magkos F, Reeds DN, Mittendorfer B. Evolution of the diagnostic value of "the sugar of the blood": hitting the sweet spot to identify alterations in glucose dynamics. Physiol Rev 2023; 103:7-30. [PMID: 35635320 PMCID: PMC9576168 DOI: 10.1152/physrev.00015.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022] Open
Abstract
In this paper, we provide an overview of the evolution of the definition of hyperglycemia during the past century and the alterations in glucose dynamics that cause fasting and postprandial hyperglycemia. We discuss how extensive mechanistic, physiological research into the factors and pathways that regulate the appearance of glucose in the circulation and its uptake and metabolism by tissues and organs has contributed knowledge that has advanced our understanding of different types of hyperglycemia, namely prediabetes and diabetes and their subtypes (impaired fasting plasma glucose, impaired glucose tolerance, combined impaired fasting plasma glucose, impaired glucose tolerance, type 1 diabetes, type 2 diabetes, gestational diabetes mellitus), their relationships with medical complications, and how to prevent and treat hyperglycemia.
Collapse
Affiliation(s)
- Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Dominic N Reeds
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
23
|
Pafili K, Kahl S, Mastrototaro L, Strassburger K, Pesta D, Herder C, Pützer J, Dewidar B, Hendlinger M, Granata C, Saatmann N, Yavas A, Gancheva S, Heilmann G, Esposito I, Schlensak M, Roden M. Mitochondrial respiration is decreased in visceral but not subcutaneous adipose tissue in obese individuals with fatty liver disease. J Hepatol 2022; 77:1504-1514. [PMID: 35988689 DOI: 10.1016/j.jhep.2022.08.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Adipose tissue dysfunction is involved in the development of insulin resistance and is responsible for excessive lipid delivery to other organs such as the liver. We tested the hypothesis that impaired mitochondrial function is a common feature of subcutaneous (SAT) and visceral adipose tissue (VAT), but may differently contribute to adipose tissue insulin resistance (IR) in obesity, non-alcoholic fatty liver (NAFL) and steatohepatitis (NASH). METHODS In this cross-sectional study, we analyzed tissue-specific insulin sensitivity using stable isotope dilution and hyperinsulinemic-normoglycemic clamp tests. We also assessed mitochondrial respiration, mRNA and protein expression, and tissue morphology in biopsies of SAT and VAT from obese humans without NAFL, with NAFL or with NASH (n = 22/group). RESULTS Compared to individuals without liver disease, persons with NAFL and NASH had about 30% (p = 0.010) and 33% (p = 0.002) lower maximal mitochondrial respiration, respectively, in VAT, but not in SAT. The lower maximal mitochondrial respiration of VAT was associated with lower adipose tissue insulin sensitivity (β = 0.985, p = 0.041) and with increased VAT protein expression of tumor necrosis factor A across all groups (β = -0.085, p = 0.040). VAT from individuals with NASH was characterized by lower expression of oxidative phosphorylation complex IV (p = 0.042) and higher mRNA expression of the macrophage marker CD68 (p = 0.002) than VAT from participants without NAFL. CONCLUSIONS Humans with non-alcoholic fatty liver disease have distinct abnormalities of VAT energy metabolism, which correlate with adipose tissue dysfunction and may favor progression of NAFL to NASH. LAY SUMMARY Adipose tissue (commonly called body fat) can be found under the skin (subcutaneous) or around internal organs (visceral). Dysfunction of adipose tissue can cause insulin resistance and lead to excess delivery of fat to other organs such as the liver. Herein, we show that dysfunction specifically in visceral adipose tissue was associated with fatty liver disease. CLINICAL TRIAL NUMBER NCT01477957.
Collapse
Affiliation(s)
- Kalliopi Pafili
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Sabine Kahl
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine-University, 40225, Düsseldorf, Germany
| | - Dominik Pesta
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Aerospace Center (DLR), Institute of Aerospace Medicine, 51147, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50931, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany
| | - Jennifer Pützer
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Bedair Dewidar
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Mona Hendlinger
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Cesare Granata
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Nina Saatmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Aslihan Yavas
- Institute of Pathology, University Hospital and Heinrich-Heine-University, 40225, Düsseldorf, Germany
| | - Sofiya Gancheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany
| | - Geronimo Heilmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany
| | - Irene Esposito
- Institute of Pathology, University Hospital and Heinrich-Heine-University, 40225, Düsseldorf, Germany
| | - Matthias Schlensak
- Department of General and Visceral Surgery, Neuwerk Hospital, 41066, Mönchengladbach, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, 85764, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
24
|
Yoshino M, Yoshino J, Smith GI, Stein RI, Bittel AJ, Bittel DC, Reeds DN, Sinacore DR, Cade WT, Patterson BW, Cho K, Patti GJ, Mittendorfer B, Klein S. Worksite-based intensive lifestyle therapy has profound cardiometabolic benefits in people with obesity and type 2 diabetes. Cell Metab 2022; 34:1431-1441.e5. [PMID: 36084645 PMCID: PMC9728552 DOI: 10.1016/j.cmet.2022.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/01/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
Lifestyle therapy (energy restriction and exercise) is the cornerstone of therapy for people with type 2 diabetes (T2D) but is difficult to implement. We conducted an 8-month randomized controlled trial in persons with obesity and T2D (17 women and 1 man) to determine the therapeutic effects and potential mechanisms of intensive lifestyle therapy on cardiometabolic function. Intensive lifestyle therapy was conducted at the worksite to enhance compliance and resulted in marked (17%) weight loss and beneficial changes in body fat mass, intrahepatic triglyceride content, cardiorespiratory fitness, muscle strength, glycemic control, β cell function, and multi-organ insulin sensitivity, which were associated with changes in muscle NAD+ biosynthesis, sirtuin signaling, and mitochondrial function and in adipose tissue remodeling. These findings demonstrate that intensive lifestyle therapy provided at the worksite has profound therapeutic clinical and physiological effects in people with T2D, which are likely mediated by specific alterations in skeletal muscle and adipose tissue biology.
Collapse
Affiliation(s)
- Mihoko Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA
| | - Jun Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA; Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Gordon I Smith
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA
| | - Richard I Stein
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA
| | - Adam J Bittel
- Program in Physical Therapy, Washington University School of Medicine, St Louis, MO, USA
| | - Daniel C Bittel
- Program in Physical Therapy, Washington University School of Medicine, St Louis, MO, USA
| | - Dominic N Reeds
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA
| | - David R Sinacore
- Program in Physical Therapy, Washington University School of Medicine, St Louis, MO, USA; Department of Physical Therapy, High Point University, High Point, NC, USA
| | - W Todd Cade
- Program in Physical Therapy, Washington University School of Medicine, St Louis, MO, USA
| | - Bruce W Patterson
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA
| | - Kevin Cho
- Department of Chemistry, Washington University School of Medicine, St Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St Louis, MO, USA; Sansum Diabetes Research Institute, Santa Barbara, CA, USA.
| |
Collapse
|
25
|
Eslam M, El-Serag HB, Francque S, Sarin SK, Wei L, Bugianesi E, George J. Metabolic (dysfunction)-associated fatty liver disease in individuals of normal weight. Nat Rev Gastroenterol Hepatol 2022; 19:638-651. [PMID: 35710982 DOI: 10.1038/s41575-022-00635-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
Metabolic (dysfunction)-associated fatty liver disease (MAFLD) affects up to a third of the global population; its burden has grown in parallel with rising rates of type 2 diabetes mellitus and obesity. MAFLD increases the risk of end-stage liver disease, hepatocellular carcinoma, death and liver transplantation and has extrahepatic consequences, including cardiometabolic disease and cancers. Although typically associated with obesity, there is accumulating evidence that not all people with overweight or obesity develop fatty liver disease. On the other hand, a considerable proportion of patients with MAFLD are of normal weight, indicating the importance of metabolic health in the pathogenesis of the disease regardless of body mass index. The clinical profile, natural history and pathophysiology of patients with so-called lean MAFLD are not well characterized. In this Review, we provide epidemiological data on this group of patients and consider overall metabolic health and metabolic adaptation as a framework to best explain the pathogenesis of MAFLD and its heterogeneity in individuals of normal weight and in those who are above normal weight. This framework provides a conceptual schema for interrogating the MAFLD phenotype in individuals of normal weight that can translate to novel approaches for diagnosis and patient care.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| | - Hashem B El-Serag
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Medicine and Paediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
26
|
GLP-1 Agonist to Treat Obesity and Prevent Cardiovascular Disease: What Have We Achieved so Far? Curr Atheroscler Rep 2022; 24:867-884. [PMID: 36044100 DOI: 10.1007/s11883-022-01062-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW To discuss evidence supporting the use of glucagon-like peptide 1 receptor agonists (GLP-1RA) to treat obesity and their role as a cardioprotective drug. Obesity is not just a hypertrophy of the adipose tissue because it may become dysfunctional and inflamed resulting in increased insulin resistance. Being overweight is associated with increased incidence of cardiovascular events and weight loss achieved through lifestyle changes lowers risk factors, but has no clear effect on cardiovascular outcomes. In contrast, treating obesity with GLP-1RA decreases cardiovascular risk and the possible mechanisms of cardioprotection achieved by this class of drugs are discussed. GLP-1RA were initially developed to treat type 2 diabetes patients, in whom the effects upon glycemia and, moreover, weight loss, especially with long-acting GLP-1RA, were evident. However, cardiovascular safety trials in type 2 diabetes patients, the majority presenting cardiovascular disease and excess weight, showed that GLP-1 receptor agonists were indeed capable of decreasing cardiovascular risk. RECENT FINDINGS Type 2 diabetes treatment with GLP-1RA liraglutide and semaglutide paved way to a ground-breaking therapy specific for obesity, as shown with the SCALE 3 mg/day liraglutide program and the STEP 2.4 mg/week semaglutide program. A novel molecule with superior performance is tirzepatide, a GLP-1 and GIP (Gastric Inhibitory Peptide) receptor agonist and recent results from the SURPASS and SURMOUNT programs are briefly described. Liraglutide was approved without a CVOT (Cardiovascular Outcome Trial) because authorities accepted the results from the LEADER study, designed for superiority. The SELECT study with semaglutide will report results only in 2023 and tirzepatide is being tested in patients with diabetes in the SURPASS-CVOT. Clinical studies highlight that GLP-1RA to treat obesity, alongside their concomitant cardioprotective effects, have become a hallmark in clinical science.
Collapse
|
27
|
Abstract
Adipose tissue is a complex heterogeneous tissue composed of adipocytes along with several non-adipocyte populations, including blood, stromal, endothelial, and progenitor cells, as well as extracellular matrix (ECM) components. As obesity progresses, the adipose tissue expands dynamically through adipocyte hypertrophy and/or hyperplasia. This expansion requires continuous ECM remodeling to properly accommodate the size increase as well as functional changes. Upon reaching a hypertrophic threshold beyond the adipocyte buffering capacity, excess ECM components are deposited, causing fibrosis and ultimately resulting in unhealthy metabolic maladaptation. These complex ECM remodeling processes in adipose tissues are regulated by the local environment, several key mediators, and genetic factors that are closely linked to insulin sensitivity. It is crucial to understand how adipocytes interact with nonadipocyte populations and various mediators (i.e., immune cells, ECM components, and adipokines) during these processes. This mini-review provides an overview of the latest research into the biology of obesity-induced adipose tissue fibrosis and its related clinical manifestations, providing insight for further studies aimed at controlling metabolic syndrome and its comorbidities.
Collapse
Affiliation(s)
- Yutaka Hasegawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
28
|
Chronic Inflammation—A Link between Nonalcoholic Fatty Liver Disease (NAFLD) and Dysfunctional Adipose Tissue. Medicina (B Aires) 2022; 58:medicina58050641. [PMID: 35630058 PMCID: PMC9147364 DOI: 10.3390/medicina58050641] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a new challenge in modern medicine, due to its high prevalence in the world. The pathogenesis of NAFLD is a complex dysmetabolic process, following the “multiple-hit” hypothesis that involves hepatocytes excessive accumulation of triglycerides, insulin resistance (IR), increased oxidative stress, chronic low-grade inflammatory response and lipotoxicity. In this review, we provide an overview of the interrelation of these processes, the link between systemic and local inflammation and the role of dysfunctional adipose tissue (AT) in the NAFLD development. Multiple extrahepatic triggers of the pathophysiological mechanisms of NAFLD are described: nutritional deficiency or malnutrition, unhealthy food intake, the dysfunction of the liver–gut axis, the involvement of the mesenteric adipose tissue, the role of adipokines such as adiponectin, of food intake hormone, the leptin and leptin resistance (LR) and adipose tissue’s hormone, the resistin. In addition, a wide range of intrahepatic players are involved: oxidative stress, fatty acid oxidation, endoplasmic reticulum stress, mitochondrial dysfunction, resident macrophages (Kupffer cells), neutrophils, dendritic cells (DCs), B and T lymphocytes contributing to the potential evolution of NAFLD to nonalcoholic steatohepatitis (NASH). This interdependent approach to complex dysmetabolic imbalance in NAFLD, integrating relevant studies, could contribute to a better clarification of pathogenesis and consequently the development of new personalized treatments, targeting de novo lipogenesis, chronic inflammation and fibrosis. Further studies are needed to focus not only on treatment, but also on prevention strategy in NAFLD.
Collapse
|
29
|
Stefan N, Cusi K. A global view of the interplay between non-alcoholic fatty liver disease and diabetes. Lancet Diabetes Endocrinol 2022; 10:284-296. [PMID: 35183303 DOI: 10.1016/s2213-8587(22)00003-1] [Citation(s) in RCA: 300] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become an epidemic, much like other non-communicable diseases (NCDs), such as cancer, obesity, diabetes, and cardiovascular disease. The pathophysiology of NAFLD, particularly involving insulin resistance and subclinical inflammation, is not only closely linked to that of those NCDs but also to a severe course of the communicable disease COVID-19. Genetics alone cannot explain the large increase in the prevalence of NAFLD during the past 2 decades and the increase that is projected for the next decades. Impairment of glucose and lipid metabolic pathways, which has been propelled by the worldwide increase in the prevalence of obesity and type 2 diabetes, is most likely behind the increase in people with NAFLD. As the prevalence of NAFLD varies among subgroups of patients with diabetes and prediabetes identified by cluster analyses, stratification of people with diabetes and prediabetes by major pathological mechanistic pathways might improve the diagnosis of NAFLD and prediction of its progression. In this Review, we aim to understand how diabetes can affect the development of hepatic steatosis and its progression to advanced liver damage. First, we emphasise the extent to which NAFLD and diabetes jointly occur worldwide. Second, we address the major mechanisms that are involved in the pathogenesis of NAFLD and type 2 diabetes, and we discuss whether these mechanisms place NAFLD in an important position to better understand the pathogenesis of NCDs and communicable diseases, such as COVID-19. Third, we address whether this knowledge can be used for personalised treatment of NAFLD in the future. Finally, we discuss the current treatment strategies for people with type 2 diabetes and their effectiveness in treating the spectrum of hepatic diseases from simple steatosis to non-alcoholic steatohepatitis and hepatic fibrosis.
Collapse
Affiliation(s)
- Norbert Stefan
- Department of Internal Medicine IV and Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University Hospital Tübingen, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany.
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| |
Collapse
|
30
|
Keating MF, Drew BG, Calkin AC. Antisense Oligonucleotide Technologies to Combat Obesity and Fatty Liver Disease. Front Physiol 2022; 13:839471. [PMID: 35295579 PMCID: PMC8918623 DOI: 10.3389/fphys.2022.839471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
Synthetic oligonucleotide technologies are DNA or RNA based molecular compounds that are utilized to disrupt gene transcription or translation in target tissues or cells. Optimally, oligonucleotides are 10–30 base pairs in length, and mediate target gene suppression through directed sequence homology with messenger RNA (mRNA), leading to mRNA degradation. Examples of specific oligonucleotide technologies include antisense oligonucleotides (ASO), short hairpin RNAs (shRNA), and small interfering RNAs (siRNA). In vitro and in vivo studies that model obesity related disorders have demonstrated that oligonucleotide technologies can be implemented to improve the metabolism of cells and tissues, exemplified by improvements in fat utilization and hepatic insulin signaling, respectively. Oligonucleotide therapy has also been associated with reductions in lipid accumulation in both the liver and adipose tissue in models of diet-induced obesity. Recent advances in oligonucleotide technologies include the addition of chemical modifications such as N-acetylgalactosamine (GalNAc) conjugates that have been successful at achieving affinity for the liver, in turn improving specificity, and thus reducing off target effects. However, some challenges are still yet to be overcome relating to hepatic injury and off-target effects that have been reported with some compounds, including ASOs. In summary, oligonucleotide-based therapies are an effective tool to elucidate mechanistic insights into metabolic pathways and provide an attractive avenue for translational research into the clinic.
Collapse
Affiliation(s)
- Michael F Keating
- Lipid Metabolism and Cardiometabolic Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Molecular Metabolism and Ageing Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Disease, University of Melbourne, Parkville, VIC, Australia
| | - Brian G Drew
- Molecular Metabolism and Ageing Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Disease, University of Melbourne, Parkville, VIC, Australia.,Central Clinical School, Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Anna C Calkin
- Lipid Metabolism and Cardiometabolic Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Disease, University of Melbourne, Parkville, VIC, Australia.,Central Clinical School, Department of Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Sarsenbayeva A, Pereira MJ, Nandi Jui B, Ahmed F, Dipta P, Fanni G, Almby K, Kristófi R, Hetty S, Eriksson JW. Excess glucocorticoid exposure contributes to adipose tissue fibrosis which involves macrophage interaction with adipose precursor cells. Biochem Pharmacol 2022; 198:114976. [PMID: 35202577 DOI: 10.1016/j.bcp.2022.114976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
Abstract
Chronic exposure to elevated glucocorticoid levels, as seen in patients with Cushing's syndrome, can induce adipose tissue fibrosis. Macrophages play a pivotal role in adipose tissue remodelling. We used the synthetic glucocorticoid analogue dexamethasone to address glucocorticoid effects on adipose tissue fibrosis, in particular involving macrophage to preadipocyte communication. We analysed the direct effects of dexamethasone at a supra-physiological level, 0.3 µM, on gene expression of pro-fibrotic markers in human subcutaneous adipose tissue. The effects of dexamethasone on the differentiation of human SGBS preadipocytes were assessed in the presence or absence of THP1-macrophages or macrophage-conditioned medium. We measured the expression of different pro-fibrotic factors, including α-smooth muscle actin gene (ACTA2) and protein (α-SMA). Dexamethasone increased the expression of pro-fibrotic genes, e.g. CTGF, COL6A3, FN1, in adipose tissue. Macrophages abolished preadipocyte differentiation and increased the expression of the ACTA2 gene and α-SMA protein in preadipocytes after differentiation. Exposure to dexamethasone during differentiation reduced adipogenesis in preadipocytes, and elevated the expression of pro-fibrotic genes. Moreover, dexamethasone added together with macrophages further increased ACTA2 and α-SMA expression in preadipocytes, making them more myofibroblast-like. Cells differentiated in the presence of conditioned media from macrophages pretreated with or without dexamethasone had a higher expression of profibrotic genes compared to control cells. Our data suggest that macrophages promote adipose tissue fibrosis by directly interfering with preadipocyte differentiation and stimulating gene expression of pro-fibrotic factors. Excess glucocorticoid exposure also has pro-fibrotic effect on adipose tissue, but this requires the presence of macrophages.
Collapse
Affiliation(s)
- Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Bipasha Nandi Jui
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Fozia Ahmed
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Priya Dipta
- Department of Pharmacology, Faculty of Medicine, Hadassah Medical Centre, Jerusalem, Israel
| | - Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Kristina Almby
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Robin Kristófi
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
32
|
Abstract
The accumulation of an excessive amount of body fat can cause type 2 diabetes, and the risk of type 2 diabetes increases linearly with an increase in body mass index. Accordingly, the worldwide increase in the prevalence of obesity has led to a concomitant increase in the prevalence of type 2 diabetes. The cellular and physiological mechanisms responsible for the link between obesity and type 2 diabetes are complex and involve adiposity-induced alterations in β cell function, adipose tissue biology, and multi-organ insulin resistance, which are often ameliorated and can even be normalized with adequate weight loss.
Collapse
Affiliation(s)
- Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO 63110, USA; Sansum Diabetes Research Institute, Santa Barbara, CA 93105, USA.
| | - Amalia Gastaldelli
- Institute of Clinical Physiology, National Research Council-CNR, Pisa 56100, Italy
| | - Hannele Yki-Järvinen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland; Department of Medicine, University of Helsinki, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Department of Cell Biology, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| |
Collapse
|
33
|
Abstract
Obesity is a chronic and progressive process affecting whole-body energy balance and is associated with comorbidities development. In addition to increased fat mass, obesity induces white adipose tissue (WAT) inflammation and fibrosis, leading to local and systemic metabolic dysfunctions, such as insulin resistance (IR). Accordingly, limiting inflammation or fibrosis deposition may improve IR and glucose homeostasis. Although no targeted therapy yet exists to slow or reverse adipose tissue fibrosis, a number of findings have clarified the underlying cellular and molecular mechanisms. In this review, we highlight adipose tissue remodeling events shown to be associated with fibrosis deposition, with a focus on adipose progenitors involved in obesity-induced healthy as well as unhealthy WAT expansion. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Geneviève Marcelin
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; ,
| | | | - Karine Clément
- INSERM, Nutrition and Obesities: Systemic Approach (NutriOmics) Research Unit, UMRS U1269, Sorbonne Université, Paris, France; , .,Nutrition Department, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|