1
|
Lauri G, Mills K, Majumder S, Capurso G. The exposome as a target for primary prevention and a tool for early detection of pancreatic cancer. Best Pract Res Clin Gastroenterol 2025; 74:101991. [PMID: 40210335 DOI: 10.1016/j.bpg.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly aggressive malignancy with limited survival due to late stage diagnosis and scarce therapeutic options. Emerging evidence highlights the role of the "exposome," which encompasses environmental, lifestyle, and metabolic exposures, as a crucial determinant in PDAC risk and a potential avenue for early detection. This review synthesizes findings on modifiable risk factors, including smoking, obesity, diabetes, diet, and alcohol consumption, and their interplay with genetic and metabolic profiles in PDAC development. Additionally, we explore cutting-edge approaches in exposomic research, such as biobanking, electronic health record analysis, and AI-driven predictive models, to identify early biomarkers and stratify high-risk populations. This integrated framework aims to inform prevention strategies and improve early detection of PDAC.
Collapse
Affiliation(s)
- Gaetano Lauri
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Krystal Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
2
|
Lorza-Gil E, Strauss OD, Ziegler E, Kansy K, Katschke MT, Rahimi G, Neuscheler D, Sandforth L, Sandforth A, Sancar G, Kaufmann B, Hartmann D, Singer S, Mihaljevic AL, Jumpertz-von Schwartzenberg R, Sbierski-Kind J, Müller TD, Birkenfeld AL, Gerst F. Incretin-responsive human pancreatic adipose tissue organoids: A functional model for fatty pancreas research. Mol Metab 2025; 91:102067. [PMID: 39549913 PMCID: PMC11625218 DOI: 10.1016/j.molmet.2024.102067] [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: 10/07/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
OBJECTIVE Infiltration of adipocytes into the pancreatic parenchyma has been linked to impaired insulin secretion in individuals with increased genetic risk of T2D and prediabetic conditions. However, the study of this ectopic fat depot has been limited by the lack of suitable in vitro models. METHODS Here, we developed a novel 3D model of functionally mature human pancreatic adipose tissue organoids by aggregating human pancreatic adipose tissue-derived stromal vascular fraction (SVF) cells into organoids and differentiating them over 19 days. RESULTS These organoids carry biological properties of the in situ pancreatic fat, presenting levels of adipogenic markers comparable to native pancreatic adipocytes and improved lipolytic and anti-lipolytic response compared to conventional 2D cultures. The organoids harbour a small population of immune cells, mimicking in vivo adipose environment. Furthermore, they express GIPR, allowing investigation of incretin effects in pancreatic fat. In accordance, GIP and the dual GLP1R/GIPR agonist tirzepatide stimulate lipolysis but had distinct effects on the expression of proinflammatory cytokines. CONCLUSIONS This novel adipose organoid model is a valuable tool to study the metabolic impact of incretin signalling in pancreatic adipose tissue, revealing potential therapeutic targets of incretins beyond islets. The donor-specific metabolic memory of these organoids enables examination of the pancreatic fat-islet crosstalk in a donor-related metabolic context.
Collapse
Affiliation(s)
- E Lorza-Gil
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.
| | - O D Strauss
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - E Ziegler
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - K Kansy
- German Center for Diabetes Research (DZD e.V.), Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - M-T Katschke
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany; M3 Research Center, University Hospital Tübingen, Tübingen, Germany
| | - G Rahimi
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - D Neuscheler
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - L Sandforth
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - A Sandforth
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - G Sancar
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - B Kaufmann
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - D Hartmann
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - S Singer
- Department of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - A L Mihaljevic
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - R Jumpertz-von Schwartzenberg
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - J Sbierski-Kind
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany; M3 Research Center, University Hospital Tübingen, Tübingen, Germany
| | - T D Müller
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich, Neuherberg, Germany; Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany
| | - A L Birkenfeld
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - F Gerst
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at University of Tübingen, Tübingen, Germany; Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Diao B, Fan Z, Zhou B, Zhan H. Crosstalk between pancreatic cancer and adipose tissue: Molecular mechanisms and therapeutic implications. Biochem Biophys Res Commun 2024; 740:151012. [PMID: 39561650 DOI: 10.1016/j.bbrc.2024.151012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
The incidence rate of pancreatic cancer, a fatal illness with a meager 5-year survival rate, has been on the rise in recent times. When individuals accumulate excessive amounts of adipose tissue, the adipose organ becomes dysfunctional due to alterations in the adipose tissue microenvironment associated with inflammation and metabolism. This phenomenon may potentially contribute to the aberrant accumulation of fat that initiates pancreatic carcinogenesis, thereby influencing the disease's progression, resistance to treatment, and metastasis. This review presents a summary of the impact of pancreatic steatosis, visceral fat, cancer-associated adipocytes and lipid diets on the advancement of pancreatic cancer, as well as the reciprocal effects of pancreatic cancer on adipose tissue. Understanding the molecular mechanisms underlying the relationship between dysfunctional adipose tissue and pancreatic cancer better may lead to the discovery of new therapeutic targets for the disease's prevention and individualized treatment. This is especially important given the rising global incidence of obesity, which will improve the pancreatic cancer treatment options that are currently insufficient.
Collapse
Affiliation(s)
- Boyu Diao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
4
|
Della Pepa G, Salamone D, Testa R, Bozzetto L, Costabile G. Intrapancreatic fat deposition and nutritional treatment: the role of various dietary approaches. Nutr Rev 2024; 82:1820-1834. [PMID: 38153345 DOI: 10.1093/nutrit/nuad159] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023] Open
Abstract
Ectopic fat accumulation in various organs and tissues, such as the liver, muscle, kidney, heart, and pancreas, is related to impaired capacity of adipose tissue to accumulate triglycerides, as a consequence of overnutrition and an unhealthy lifestyle. Ectopic fat promotes organ dysfunction and is a key factor in the development and progression of cardiometabolic diseases. Interest in intrapancreatic fat deposition (IPFD) has developed in the last few years, particularly in relation to improvement in methodological techniques for detection of fat in the pancreas, and to growing evidence for the role that IPFD might have in glucose metabolism disorders and cardiometabolic disease. Body weight reduction represents the main option for reducing fat, and the evidence consistently shows that hypocaloric diets are effective in reducing IPFD. Changes in diet composition, independently of changes in energy intake, might offer a more feasible and safe alternative treatment to energy restriction. This current narrative review focused particularly on the possible beneficial role of the diet and its nutrient content, in hypocaloric and isocaloric conditions, in reducing IPFD in individuals with high cardiometabolic risk, highlighting the possible effects of differences in calorie quantity and calorie quality. This review also describes plausible mechanisms by which the various dietary approaches could modulate IPFD.
Collapse
Affiliation(s)
- Giuseppe Della Pepa
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
| | - Dominic Salamone
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Roberta Testa
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Lutgarda Bozzetto
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Giuseppina Costabile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| |
Collapse
|
5
|
Mehl F, Sánchez-Archidona AR, Meitil I, Gerl M, Cruciani-Guglielmacci C, Wigger L, Le Stunff H, Meneyrol K, Lallement J, Denom J, Klose C, Simons K, Pagni M, Magnan C, Ibberson M, Thorens B. A multiorgan map of metabolic, signaling, and inflammatory pathways that coordinately control fasting glycemia in mice. iScience 2024; 27:111134. [PMID: 39507247 PMCID: PMC11539597 DOI: 10.1016/j.isci.2024.111134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/07/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
To identify the pathways that are coordinately regulated in pancreatic β cells, muscle, liver, and fat to control fasting glycemia we fed C57Bl/6, DBA/2, and Balb/c mice a regular chow or a high fat diet for 5, 13, and 33 days. Physiological, transcriptomic and lipidomic data were used in a data fusion approach to identify organ-specific pathways linked to fasting glycemia across all conditions investigated. In pancreatic islets, constant insulinemia despite higher glycemic levels was associated with reduced expression of hormone and neurotransmitter receptors, OXPHOS, cadherins, integrins, and gap junction mRNAs. Higher glycemia and insulin resistance were associated, in muscle, with decreased insulin signaling, glycolytic, Krebs' cycle, OXPHOS, and endo/exocytosis mRNAs; in hepatocytes, with reduced insulin signaling, branched chain amino acid catabolism and OXPHOS mRNAs; in adipose tissue, with increased innate immunity and lipid catabolism mRNAs. These data provide a resource for further studies of interorgan communication in glucose homeostasis.
Collapse
Affiliation(s)
- Florence Mehl
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | - Ana Rodríguez Sánchez-Archidona
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Ida Meitil
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | | | | | - Leonore Wigger
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | - Hervé Le Stunff
- Université de Paris Cité, BFA, UMR 8251, CNRS, 75013 Paris, France
| | - Kelly Meneyrol
- Université de Paris Cité, BFA, UMR 8251, CNRS, 75013 Paris, France
| | | | - Jessica Denom
- Université de Paris Cité, BFA, UMR 8251, CNRS, 75013 Paris, France
| | | | | | - Marco Pagni
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | | | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | - Bernard Thorens
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
6
|
Zhang Y, Qian B, Yang Y, Niu F, Lin C, Yuan H, Wang J, Wu T, Shao Y, Shao S, Liu A, Wu J, Sun P, Chang X, Bi Y, Tang W, Zhu Y, Chen F, Su D, Han X. Visceral Adipocyte-Derived Extracellular Vesicle miR-27a-5p Elicits Glucose Intolerance by Inhibiting Pancreatic β-Cell Insulin Secretion. Diabetes 2024; 73:1832-1847. [PMID: 39186314 PMCID: PMC11493764 DOI: 10.2337/db24-0177] [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: 03/03/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Pancreatic β-cell dysfunction caused by obesity can be associated with alterations in the levels of miRNAs. However, the role of miRNAs in such processes remains elusive. Here, we show that pancreatic islet miR-27a-5p, which is markedly increased in obese mice and impairs insulin secretion, is mainly delivered by visceral adipocyte-derived extracellular vesicles (EVs). Depleting miR-27a-5p significantly improved insulin secretion and glucose intolerance in db/db mice. Supporting the function of EV miR-27a-5p as a key pathogenic factor, intravenous injection of miR-27a-5p-containing EVs showed their distribution in mouse pancreatic islets. Tracing the injected adeno-associated virus (AAV)-miR-27a-5p (AAV-miR-27a) or AAV-FABP4-miR-27a-5p (AAV-FABP4-miR-27a) in visceral fat resulted in upregulating miR-27a-5p in EVs and serum and elicited mouse pancreatic β-cell dysfunction. Mechanistically, miR-27a-5p directly targeted L-type Ca2+ channel subtype CaV1.2 (Cacna1c) and reduced insulin secretion in β-cells. Overexpressing mouse CaV1.2 largely abolished the insulin secretion injury induced by miR-27a-5p. These findings reveal a causative role of EV miR-27a-5p in visceral adipocyte-mediated pancreatic β-cell dysfunction in obesity-associated type 2 diabetes mellitus. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bin Qian
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yang Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Fandi Niu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, Xi’an, Shanxi, China
| | - Changsong Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Honglei Yuan
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianan Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yixue Shao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shulin Shao
- Department of Laboratory, Nanjing Pukou Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Aiming Liu
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingwen Wu
- The First Clinical School of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, Jiangsu, China
| | - Wei Tang
- Department of Endocrinology, Islet Cell Senescence and Function Research Laboratory, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Wang J, Wei Z, Wang Y, Zhang D, Wang M, Chen X, Xia P, Wang J, Xie C, Chen X. Pancreatic fat infiltration is associated with risk of vertebral fracture in older patients with type 2 diabetes: A longitudinal multicenter study. Diabetes Res Clin Pract 2024; 217:111904. [PMID: 39447678 DOI: 10.1016/j.diabres.2024.111904] [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: 07/29/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
AIMS Patients with type 2diabetesmellitus (T2DM) have high fracture risk. This study explored the associations between pancreatic computed tomography (CT) attenuation, a marker of pancreatic fat, and risk of vertebral fracture in T2DM patients. METHODS A total of 1486 T2DM patients who aged 50 years and older and without preexisting vertebral fractures during 2019-2023 at our institutions were followed up untilJanuary 2024. CT attenuation of the pancreas, bone and spleen were measured. Pancreatic attenuation/spleen attenuation ratio (P/S) was calculated. Vertebral fractures were evaluated on spine CT images according to Genant's semiquantitative scoring system. RESULTS A total of 135 cases of vertebral fracture were identified during 26 months of follow-up and 270 patients without vertebral fracture were matched. Pancreatic CT attenuation and the P/S ratio were negatively associated with the risk of vertebral fracture (adjusted hazard ratio (aHR) = 0.97, 95 %confidence interval (CI): 0.96-0.99; aHR = 0.26, 95 %CI: 0.12-0.58). Addition of pancreatic attenuation or P/S ratio improved the performance of bone attenuation-based model (area under the curve = 0.72-0.763 vs 0.63-0.728). CONCLUSION Pancreatic fat infiltration is an associated factor for vertebral fracture in T2DM patients. Addition of pancreatic fat infiltration improved the predictive performance of the bone-based model.
Collapse
Affiliation(s)
- Jiangchuan Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Zicheng Wei
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Dingzhe Zhang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Miaomiao Wang
- Department of Radiology, the Second Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xin Chen
- Department of Radiology, Shanghai Longhua Hospital, Shanghai 200032, China
| | - Peng Xia
- Department of Radiology, Wuxi Hospital of Traditional Chinese Medicine, Wuxi 214072, China
| | - Jianhua Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China.
| | - Chao Xie
- Centerfor Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, NY 14642, USA
| | - Xiao Chen
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
8
|
Lu X, Xie Q, Pan X, Zhang R, Zhang X, Peng G, Zhang Y, Shen S, Tong N. Type 2 diabetes mellitus in adults: pathogenesis, prevention and therapy. Signal Transduct Target Ther 2024; 9:262. [PMID: 39353925 PMCID: PMC11445387 DOI: 10.1038/s41392-024-01951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/21/2024] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
Type 2 diabetes (T2D) is a disease characterized by heterogeneously progressive loss of islet β cell insulin secretion usually occurring after the presence of insulin resistance (IR) and it is one component of metabolic syndrome (MS), and we named it metabolic dysfunction syndrome (MDS). The pathogenesis of T2D is not fully understood, with IR and β cell dysfunction playing central roles in its pathophysiology. Dyslipidemia, hyperglycemia, along with other metabolic disorders, results in IR and/or islet β cell dysfunction via some shared pathways, such as inflammation, endoplasmic reticulum stress (ERS), oxidative stress, and ectopic lipid deposition. There is currently no cure for T2D, but it can be prevented or in remission by lifestyle intervention and/or some medication. If prevention fails, holistic and personalized management should be taken as soon as possible through timely detection and diagnosis, considering target organ protection, comorbidities, treatment goals, and other factors in reality. T2D is often accompanied by other components of MDS, such as preobesity/obesity, metabolic dysfunction associated steatotic liver disease, dyslipidemia, which usually occurs before it, and they are considered as the upstream diseases of T2D. It is more appropriate to call "diabetic complications" as "MDS-related target organ damage (TOD)", since their development involves not only hyperglycemia but also other metabolic disorders of MDS, promoting an up-to-date management philosophy. In this review, we aim to summarize the underlying mechanism, screening, diagnosis, prevention, and treatment of T2D, especially regarding the personalized selection of hypoglycemic agents and holistic management based on the concept of "MDS-related TOD".
Collapse
Affiliation(s)
- Xi Lu
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Qingxing Xie
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohui Pan
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Ruining Zhang
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Zhang
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Ge Peng
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Yuwei Zhang
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Sumin Shen
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Research Centre for Diabetes and Metabolism, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
10
|
Pang C, Dong P, Yang J, Fan Z, Cheng Z, Zhan H. Non-alcoholic fatty pancreas disease: an updated review. JOURNAL OF PANCREATOLOGY 2024; 7:212-221. [DOI: 10.1097/jp9.0000000000000157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Ectopic accumulation of fat can cause a variety of metabolic diseases, and the emerging non-alcoholic fatty pancreas disease (NAFPD) is increasingly being recognized by clinicians as a cause for concern. NAFPD is a disease caused by abnormal accumulation of adipose tissue in the pancreas, which is related to obesity. The main feature of NAFPD is death of acinar cells, which are then replaced by adipose cells. However, the underlying molecular mechanisms have not been fully explored. Obesity, aging, and metabolic syndrome are independent risk factors for the occurrence and development of NAFPD. Studies have shown that NAFPD leads to insulin resistance and pancreatic dysfunction, increases the risk of diabetes mellitus, worsens the severity of pancreatitis, and is significantly correlated with pancreatic cancer and postoperative pancreatic fistula. There is no standard treatment for NAFPD; exercise, a balanced diet, and lifestyle can help reduce pancreatic fat; however, other treatment modalities such as drugs and bariatric surgery are still being explored. The specific pathological mechanism of NAFPD remains unclear, and its potential association with various clinical diseases requires further study. This review summarizes the etiology, diagnosis, clinical consequences, and potential therapeutic strategies of NAFPD.
Collapse
Affiliation(s)
- Chaoyu Pang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Peng Dong
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Jian Yang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Zhiqiang Cheng
- Division of Colorectal Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| |
Collapse
|
11
|
Sancar G, Birkenfeld AL. The role of adipose tissue dysfunction in hepatic insulin resistance and T2D. J Endocrinol 2024; 262:e240115. [PMID: 38967989 PMCID: PMC11378142 DOI: 10.1530/joe-24-0115] [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: 04/20/2024] [Accepted: 07/05/2024] [Indexed: 07/07/2024]
Abstract
The root cause of type 2 diabetes (T2D) is insulin resistance (IR), defined by the failure of cells to respond to circulating insulin to maintain lipid and glucose homeostasis. While the causes of whole-body insulin resistance are multifactorial, a major contributing factor is dysregulation of liver and adipose tissue function. Adipose dysfunction, particularly adipose tissue-IR (adipo-IR), plays a crucial role in the development of hepatic insulin resistance and the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) in the context of T2D. In this review, we will focus on molecular mechanisms of hepatic insulin resistance and its association with adipose tissue function. A deeper understanding of the pathophysiological mechanisms of the transition from a healthy state to insulin resistance, impaired glucose tolerance, and T2D may enable us to prevent and intervene in the progression to T2D.
Collapse
Affiliation(s)
- Gencer Sancar
- German Center for Diabetes Research, Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research, Neuherberg, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Kearns ML, Reynolds CM. Developmentally programmed obesity: Is there a role for anti-inflammatory nutritional strategies? Exp Physiol 2024; 109:633-646. [PMID: 38031876 PMCID: PMC11061634 DOI: 10.1113/ep091209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023]
Abstract
Pregnancy represents a period of immense maternal physiological adaptation, with progressive increases in lipid storage potential and insulin resistance to support fetal/placental growth. This requires significant change in the adipose tissue. Women living with obesity/overweight are more susceptible to these changes causing complications such as gestational diabetes. This is particularly worrying as up to 60% of European women are living with overweight/obesity at the onset of pregnancy. Furthermore, less than 1% meet all nutrition guidelines. There is now evidence that these deep metabolic changes can result in a predisposition to metabolic disease in both the mother and child in later life. Health and nutrition status during this period therefore represents a window to future health. This period offers a valuable opportunity for intervention to prevent the negative consequences of poor in utero environments and increases the long-term quality of life for mother and offspring. This review will examine a range of in utero factors which determine adipose tissue development, the impact of these factors on later-life obesity and metabolic health and the therapeutic value of dietary anti-inflammatory nutritional interventions during pregnancy and early life. When it comes to early life nutrition, a 'one size fits all' approach is not always appropriate. Understanding the mechanisms of adipose tissue development in response to differing nutritional strategies may be important in the context of complicated or adverse in utero environments and represents a substantial step towards a more personalised nutritional approach for the prevention of obesity, metabolic syndrome and related non-communicable diseases in future generations.
Collapse
Affiliation(s)
- Michelle L. Kearns
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| | - Clare M. Reynolds
- Conway Institute/School of Public Health Physiotherapy and Sports Science/Institute of Food and Health/Diabetes Complications Research CentreUniversity College DublinDublin 4Ireland
| |
Collapse
|
13
|
Taneera J, Saber-Ayad MM. Preservation of β-Cells as a Therapeutic Strategy for Diabetes. Horm Metab Res 2024; 56:261-271. [PMID: 38387480 DOI: 10.1055/a-2239-2668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The preservation of pancreatic islet β-cells is crucial in diabetes mellitus, encompassing both type 1 and type 2 diabetes. β-cell dysfunction, reduced mass, and apoptosis are central to insufficient insulin secretion in both types. Research is focused on understanding β-cell characteristics and the factors regulating their function to develop novel therapeutic approaches. In type 1 diabetes (T1D), β-cell destruction by the immune system calls for exploring immunosuppressive therapies, non-steroidal anti-inflammatory drugs, and leukotriene antagonists. Islet transplantation, stem cell therapy, and xenogeneic transplantation offer promising strategies for type 1 diabetes treatment. For type 2 diabetes (T2D), lifestyle changes like weight loss and exercise enhance insulin sensitivity and maintain β-cell function. Additionally, various pharmacological approaches, such as cytokine inhibitors and protein kinase inhibitors, are being investigated to protect β-cells from inflammation and glucotoxicity. Bariatric surgery emerges as an effective treatment for obesity and T2D by promoting β-cell survival and function. It improves insulin sensitivity, modulates gut hormones, and expands β-cell mass, leading to diabetes remission and better glycemic control. In conclusion, preserving β-cells offers a promising approach to managing both types of diabetes. By combining lifestyle modifications, targeted pharmacological interventions, and advanced therapies like stem cell transplantation and bariatric surgery, we have a significant chance to preserve β-cell function and enhance glucose regulation in diabetic patients.
Collapse
Affiliation(s)
- Jalal Taneera
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Maha M Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
14
|
Villaca CBP, Mastracci TL. Pancreatic Crosstalk in the Disease Setting: Understanding the Impact of Exocrine Disease on Endocrine Function. Compr Physiol 2024; 14:5371-5387. [PMID: 39109973 PMCID: PMC11425433 DOI: 10.1002/cphy.c230008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
The exocrine and endocrine are functionally distinct compartments of the pancreas that have traditionally been studied as separate entities. However, studies of embryonic development, adult physiology, and disease pathogenesis suggest there may be critical communication between exocrine and endocrine cells. In fact, the incidence of the endocrine disease diabetes secondary to exocrine disease/dysfunction ranges from 25% to 80%, depending on the type and severity of the exocrine pathology. Therefore, it is necessary to investigate how exocrine-endocrine "crosstalk" may impact pancreatic function. In this article, we discuss common exocrine diseases, including cystic fibrosis, acute, hereditary, and chronic pancreatitis, and the impact of these exocrine diseases on endocrine function. Additionally, we review how obesity and fatty pancreas influence exocrine function and the impact on cellular communication between the exocrine and endocrine compartments. Interestingly, in all pathologies, there is evidence that signals from the exocrine disease contribute to endocrine dysfunction and the progression to diabetes. Continued research efforts to identify the mechanisms that underlie the crosstalk between various cell types in the pancreas are critical to understanding normal pancreatic physiology as well as disease states. © 2024 American Physiological Society. Compr Physiol 14:5371-5387, 2024.
Collapse
Affiliation(s)
| | - Teresa L Mastracci
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
15
|
Yamazaki H, Streicher SA, Wu L, Fukuhara S, Wagner R, Heni M, Grossman SR, Lenz HJ, Setiawan VW, Le Marchand L, Huang BZ. Evidence for a causal link between intra-pancreatic fat deposition and pancreatic cancer: A prospective cohort and Mendelian randomization study. Cell Rep Med 2024; 5:101391. [PMID: 38280379 PMCID: PMC10897551 DOI: 10.1016/j.xcrm.2023.101391] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/24/2023] [Accepted: 12/29/2023] [Indexed: 01/29/2024]
Abstract
Prior observational studies suggest an association between intra-pancreatic fat deposition (IPFD) and pancreatic ductal adenocarcinoma (PDAC); however, the causal relationship is unclear. To elucidate causality, we conduct a prospective observational study using magnetic resonance imaging (MRI)-measured IPFD data and also perform a Mendelian randomization study using genetic instruments for IPFD. In the observational study, we use UK Biobank data (N = 29,463, median follow-up: 4.5 years) and find that high IPFD (>10%) is associated with PDAC risk (adjusted hazard ratio [HR]: 3.35, 95% confidence interval [95% CI]: 1.60-7.00). In the Mendelian randomization study, we leverage eight out of nine IPFD-associated genetic variants (p < 5 × 10-8) from a genome-wide association study in the UK Biobank (N = 25,617) and find that genetically determined IPFD is associated with PDAC (odds ratio [OR] per 1-standard deviation [SD] increase in IPFD: 2.46, 95% CI: 1.38-4.40) in the Pancreatic Cancer Cohort Consortium I, II, III (PanScan I-III)/Pancreatic Cancer Case-Control Consortium (PanC4) dataset (8,275 PDAC cases and 6,723 non-cases). This study provides evidence for a potential causal role of IPFD in the pathogenesis of PDAC. Thus, reducing IPFD may lower PDAC risk.
Collapse
Affiliation(s)
- Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Center for Innovative Research for Communities and Clinical Excellence (CiRC2LE), Fukushima Medical University, Fukushima, Japan.
| | - Samantha A Streicher
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Shunichi Fukuhara
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Center for Innovative Research for Communities and Clinical Excellence (CiRC2LE), Fukushima Medical University, Fukushima, Japan; Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Róbert Wagner
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Heni
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, Ulm University, Ulm, Germany; Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Steven R Grossman
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Heinz-Josef Lenz
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Veronica Wendy Setiawan
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Loïc Le Marchand
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Brian Z Huang
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
16
|
An Q, Zhang QH, Wang Y, Zhang HY, Liu YH, Zhang ZT, Zhang ML, Lin LJ, He H, Yang YF, Sun P, Zhou ZY, Song QW, Liu AL. Association between type 2 diabetes mellitus and body composition based on MRI fat fraction mapping. Front Public Health 2024; 12:1332346. [PMID: 38322122 PMCID: PMC10846073 DOI: 10.3389/fpubh.2024.1332346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Purpose To explore the association between type 2 diabetes mellitus (T2DM) and body composition based on magnetic resonance fat fraction (FF) mapping. Methods A total of 341 subjects, who underwent abdominal MRI examination with FF mapping were enrolled in this study, including 68 T2DM patients and 273 non-T2DM patients. The FFs and areas of visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT) and abdominal muscle (AM) were measured at the level of the L1-L2 vertebral. The FF of bone marrow adipose tissue (BMAT) was determined by the averaged FF values measured at the level of T12 and L1 vertebral, respectively. The whole hepatic fat fraction (HFF) and pancreatic fat fraction (PFF) were measured based on 3D semi-automatic segmentation on the FF mapping. All data were analyzed by GraphPad Prism and MedCalc. Results VAT area, VAT FF, HFF, PFF of T2DM group were higher than those of non-T2DM group after adjusting for age and sex (P < 0.05). However, there was no differences in SAT area, SAT FF, BMAT FF, AM area and AM FF between the two groups (P > 0.05). VAT area and PFF were independent risk factors of T2DM (all P < 0.05). The area under the curve (AUC) of the receiver operating characteristic (ROC) for VAT area and PFF in differentiating between T2DM and non-T2DM were 0.685 and 0.787, respectively, and the AUC of PFF was higher than VAT area (P < 0.05). Additionally, in seemingly healthy individuals, the SAT area, VAT area, and AM area were found to be significantly associated with being overweight and/or obese (BMI ≥ 25) (all P < 0.05). Conclusions In this study, it was found that there were significant associations between T2DM and VAT area, VAT FF, HFF and PFF. In addition, VAT area and PFF were the independent risk factors of T2DM. Especially, PFF showed a high diagnostic performance in discrimination between T2DM and non-T2DM. These findings may highlight the crucial role of PFF in the pathophysiology of T2DM, and it might be served as a potential imaging biomarker of the prevention and treatment of T2DM. Additionally, in individuals without diabetes, focusing on SAT area, VAT area and AM area may help identify potential health risks and provide a basis for targeted weight management and prevention measures.
Collapse
Affiliation(s)
- Qi An
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qin-He Zhang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Han-Yue Zhang
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu-Hui Liu
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | - Zi-Ting Zhang
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | - Mei-Ling Zhang
- Department of Medical Imaging, Dalian Medical University, Dalian, China
| | | | - Hui He
- Department of Thyroid, Metabolic Diseases and Hernia Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yi-Fan Yang
- Department of Thyroid, Metabolic Diseases and Hernia Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Peng Sun
- Philips Healthcare, Beijing, China
| | | | - Qing-Wei Song
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ai-Lian Liu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Xourafa G, Korbmacher M, Roden M. Inter-organ crosstalk during development and progression of type 2 diabetes mellitus. Nat Rev Endocrinol 2024; 20:27-49. [PMID: 37845351 DOI: 10.1038/s41574-023-00898-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2023] [Indexed: 10/18/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by tissue-specific insulin resistance and pancreatic β-cell dysfunction, which result from the interplay of local abnormalities within different tissues and systemic dysregulation of tissue crosstalk. The main local mechanisms comprise metabolic (lipid) signalling, altered mitochondrial metabolism with oxidative stress, endoplasmic reticulum stress and local inflammation. While the role of endocrine dysregulation in T2DM pathogenesis is well established, other forms of inter-organ crosstalk deserve closer investigation to better understand the multifactorial transition from normoglycaemia to hyperglycaemia. This narrative Review addresses the impact of certain tissue-specific messenger systems, such as metabolites, peptides and proteins and microRNAs, their secretion patterns and possible alternative transport mechanisms, such as extracellular vesicles (exosomes). The focus is on the effects of these messengers on distant organs during the development of T2DM and progression to its complications. Starting from the adipose tissue as a major organ relevant to T2DM pathophysiology, the discussion is expanded to other key tissues, such as skeletal muscle, liver, the endocrine pancreas and the intestine. Subsequently, this Review also sheds light on the potential of multimarker panels derived from these biomarkers and related multi-omics for the prediction of risk and progression of T2DM, novel diabetes mellitus subtypes and/or endotypes and T2DM-related complications.
Collapse
Affiliation(s)
- Georgia Xourafa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Düsseldorf, Germany
| | - Melis Korbmacher
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Düsseldorf, Germany.
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
18
|
Mathrani A, Yip W, Sequeira-Bisson IR, Barnett D, Stevenson O, Taylor MW, Poppitt SD. Effect of a 12-Week Polyphenol Rutin Intervention on Markers of Pancreatic β-Cell Function and Gut Microbiota in Adults with Overweight without Diabetes. Nutrients 2023; 15:3360. [PMID: 37571297 PMCID: PMC10420824 DOI: 10.3390/nu15153360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Supplementation with prebiotic polyphenol rutin is a potential dietary therapy for type 2 diabetes prevention in adults with obesity, based on previous glycaemic improvement in transgenic mouse models. Gut microbiota are hypothesised to underpin these effects. We investigated the effect of rutin supplementation on pancreatic β-cell function measured as C-peptide/glucose ratio, and 16S rRNA gene-based gut microbiota profiles, in a cohort of individuals with overweight plus normoglycaemia or prediabetes. Eighty-seven participants were enrolled, aged 18-65 years with BMI of 23-35 kg/m2. This was a 12-week double-blind randomised controlled trial (RCT), with 3 treatments comprising (i) placebo control, (ii) 500 mg/day encapsulated rutin, and (iii) 500 mg/day rutin-supplemented yoghurt. A 2-h oral glucose tolerance test (OGTT) was performed at baseline and at the end of the trial, with faecal samples also collected. Compliance with treatment was high (~90%), but rutin in both capsule and dietary format did not alter pancreatic β-cell response to OGTT over 12 weeks. Gut bacterial community composition also did not significantly change, with Firmicutes dominating irrespective of treatment. Fasting plasma glucose negatively correlated with the abundance of the butyrate producer Roseburia inulinivorans, known for its anti-inflammatory capacity. This is the first RCT to investigate postprandial pancreatic β-cell function in response to rutin supplementation.
Collapse
Affiliation(s)
- Akarsh Mathrani
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand; (A.M.); (W.Y.); (I.R.S.-B.)
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
| | - Wilson Yip
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand; (A.M.); (W.Y.); (I.R.S.-B.)
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
- Human Nutrition Unit, University of Auckland, Auckland 1024, New Zealand
| | - Ivana R. Sequeira-Bisson
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand; (A.M.); (W.Y.); (I.R.S.-B.)
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
- Human Nutrition Unit, University of Auckland, Auckland 1024, New Zealand
| | - Daniel Barnett
- Department of Statistics, University of Auckland, Auckland 1010, New Zealand; (D.B.); (O.S.)
| | - Oliver Stevenson
- Department of Statistics, University of Auckland, Auckland 1010, New Zealand; (D.B.); (O.S.)
| | - Michael W. Taylor
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand; (A.M.); (W.Y.); (I.R.S.-B.)
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
| | - Sally D. Poppitt
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand; (A.M.); (W.Y.); (I.R.S.-B.)
- High-Value Nutrition National Science Challenge, Auckland 1010, New Zealand
- Human Nutrition Unit, University of Auckland, Auckland 1024, New Zealand
- Department of Medicine, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
19
|
Källner K, Krook R, Sandberg AS, Hulthén L, Andersson-Hall U, Holmäng A. Interaction of Iron Homeostasis and Fatty Acid Metabolism in the Development of Glucose Intolerance in Women with Previous Gestational Diabetes Mellitus. Nutrients 2023; 15:3214. [PMID: 37513632 PMCID: PMC10385184 DOI: 10.3390/nu15143214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
A gestational diabetes mellitus (GDM) diagnosis during pregnancy means an increased risk of developing type 2 diabetes later in life. By following up with women after GDM we aimed to examine the relationship between iron parameters, individual fatty acids (FAs) and desaturases in the development of impaired glucose metabolism (IGM). Based on an oral glucose tolerance test (OGTT), six years after GDM, 157 women were grouped as having normal glucose tolerance (NGT) or IGM. Fasting serum FAs, activity of desaturases and iron parameters (ferritin, transferrin, iron, soluble transferrin receptor, total iron binding capacity, hepcidin) were measured, and clinical and anthropometric measurements taken. Soluble transferrin receptor was higher in the IGM group compared to the NGT group (3.87 vs. 3.29 mg/L, p-value = 0.023) and associated positively with saturated FAs and negatively with monounsaturated FAs in the IGM group (adjusted for BMI, age and high sensitivity C-reactive protein; p-value < 0.05). Iron, as well as transferrin saturation, showed a positive association with MUFAs and desaturase activity. These associations were not seen in the NGT group. These results suggest that iron homeostasis and FA metabolism interact in the development of glucose intolerance in women with previous GDM.
Collapse
Affiliation(s)
- Kristin Källner
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden (U.A.-H.)
| | - Rasmus Krook
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden (U.A.-H.)
| | - Ann-Sofie Sandberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Lena Hulthén
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Ulrika Andersson-Hall
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden (U.A.-H.)
| | - Agneta Holmäng
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden (U.A.-H.)
| |
Collapse
|
20
|
Bocian-Jastrzębska A, Malczewska-Herman A, Rosiek V, Kos-Kudła B. Assessment of the Role of Leptin and Adiponectinas Biomarkers in Pancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2023; 15:3517. [PMID: 37444627 DOI: 10.3390/cancers15133517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Data on the possible connection between circulating adipokines and PanNENs are limited. This novel study aimed to assess the serum levels of leptin and adiponectin and their ratio in patients with PanNENs and to evaluate the possible relationship between them and PanNEN's grade or stage, including the presence of metastases. The study group consisted of PanNENs (n = 83), and healthy controls (n = 39). Leptin and adiponectin measurement by an ELISA assay was undertaken in the entire cohort. The serum concentration of adiponectin was significantly higher in the control group compared to the study group (p < 0.001). The concentration of leptin and adiponectin was significantly higher in females than in males (p < 0.01). Anincreased leptin-adiponectin ratio was observed in well-differentiated PanNENs (G1) vs. moderatelydifferentiated PanNENs (G2) (p < 0.05). An increased leptin-adiponectin ratio was found in PanNENs with Ki-67 < 3% vs. Ki-67 ≥ 3% (p < 0.05). PanNENs with distal disease presented lower leptin levels (p < 0.001) and a decreased leptin-adiponectin ratio (p < 0.01) compared with the localized disease group. Leptin, adiponectin, and the leptin-adiponectin ratio may serve as potential diagnostic, prognostic, and predictive biomarkers for PanNENs. Leptin levels and the leptin-adiponectin ratio may play an important role as predictors of malignancy and metastasis in PanNENs.
Collapse
Affiliation(s)
- Agnes Bocian-Jastrzębska
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinogy, Medical University of Silesia, 40-514 Katowice, Poland
| | - Anna Malczewska-Herman
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinogy, Medical University of Silesia, 40-514 Katowice, Poland
| | - Violetta Rosiek
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinogy, Medical University of Silesia, 40-514 Katowice, Poland
| | - Beata Kos-Kudła
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinogy, Medical University of Silesia, 40-514 Katowice, Poland
| |
Collapse
|
21
|
Siegel-Axel D, Barroso Oquendo M, Gerst F, Fend F, Wagner R, Heni M, Königsrainer A, Häring HU, Fritsche A, Schleicher E, Birkenfeld AL, Stefan N. Extracellular Matrix Expression in Human Pancreatic Fat Cells of Patients with Normal Glucose Regulation, Prediabetes and Type 2 Diabetes. Int J Mol Sci 2023; 24:11169. [PMID: 37446346 DOI: 10.3390/ijms241311169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Previously, we found that human pancreatic preadipocytes (PPAs) and islets influence each other and that the crosstalk with the fatty liver via the hepatokine fetuin-A/palmitate induces inflammatory responses. Here, we examined whether the mRNA-expression of pancreatic extracellular matrix (ECM)-forming and -degrading components differ in PPAs from individuals with normal glucose regulation (PPAs-NGR), prediabetes (PPAs-PD), and type 2 diabetes (PPAs-T2D), and whether fetuin-A/palmitate impacts ECM-formation/degradation and associated monocyte invasion. Human pancreatic resections were analyzed (immuno)histologically. PPAs were studied for mRNA expression by real-time PCR and protein secretion by Luminex analysis. Furthermore, co-cultures with human islets and monocyte migration assays in Transwell plates were conducted. We found that in comparison with NGR-PPAs, TIMP-2 mRNA levels were lower in PPAs-PD, and TGF-β1 mRNA levels were higher in PPAs-T2D. Fetuin-A/palmitate reduced fibronectin, decorin, TIMP-1/-2 and TGF-ß1 mRNA levels. Only fibronectin was strongly downregulated by fetuin-A/palmitate independently of the glycemic status. Co-culturing of PPAs with islets increased TIMP-1 mRNA expression in islets. Fetuin-A/palmitate increased MMP-1, usherin and dermatopontin mRNA-levels in co-cultured islets. A transmigration assay showed increased monocyte migration towards PPAs, which was enhanced by fetuin-A/palmitate. This was more pronounced in PPAs-T2D. The expression of distinct ECM components differs in PPAs-PD and PPAs-T2D compared to PPAs-NGR, suggesting that ECM alterations can occur even in mild hyperglycemia. Fetuin-A/palmitate impacts on ECM formation/degradation in PPAs and co-cultured islets. Fetuin-A/palmitate also enhances monocyte migration, a process which might impact on matrix turnover.
Collapse
Affiliation(s)
- Dorothea Siegel-Axel
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Morgana Barroso Oquendo
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- EKU Tübingen, Quantitative Biology Center (QBiC), University of Tübingen, 72076 Tübingen, Germany
| | - Felicia Gerst
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Falko Fend
- Department of General Pathology and Pathological Anatomy, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Robert Wagner
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Heinrich Heine University Düsseldorf (HHU), 40225 Düsseldorf, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital Ulm, 89081 Ulm, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Erwin Schleicher
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| |
Collapse
|
22
|
Yang H, Zhang M, Nie J, Zhang M, Lu G, Chen R, He Q. Associations of obesity-related indices with prediabetes regression to normoglycemia among Chinese middle-aged and older adults: a prospective study. Front Nutr 2023; 10:1075225. [PMID: 37275653 PMCID: PMC10235473 DOI: 10.3389/fnut.2023.1075225] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/26/2023] [Indexed: 06/07/2023] Open
Abstract
Background Prediabetes is associated with increased cardiovascular risk and all-cause mortality, while its regression will decrease the risks. This study investigated the associations of six obesity-related indices (waist-to-height ratio (WHtR), body roundness index (BRI), conicity index (CI), body shape index (ABSI), Chinese visceral adiposity index (CVAI), and triglyceride glucose (TyG) index) with prediabetes regression based on the China Health and Retirement Longitudinal Study (CHARLS), enrolling middle-aged and older adults. Methods We included 2,601 participants with prediabetes from CHARLS, who were followed up from 2011-2012 to 2015-2016, with blood samples collected for measuring fasting plasma glucose and hemoglobin A1c. All the obesity-related indices at baseline and their dynamic changes were calculated and categorized into tertiles. Logistic regression analysis was applied to obtain the odds ratio (OR) and 95% confidence intervals (CIs). Attributable fractions (AFs) and 95% CIs of these indices and the dynamic changes were calculated with the AF package in R software, and the cutoff values of initial obesity-related indices were obtained by the receiver operating characteristic (ROC) analysis. Results During the 4-year follow-up period, 562 (21.61%) participants regressed from prediabetes to normoglycemia. They had lower initial BRI, WHtR, CI, ABSI, CVAI, and TyG than those who did not (P < 0.05). After multivariable adjustment, participants in the first tertile of initial BRI (OR, 1.45, 95%CIs, 1.09-1.93), WHtR (OR, 1.46, 95%CIs, 1.10-1.95), and CVAI (OR, 1.47, 95%CIs, 1.11-1.93) had increased odds of prediabetes regression compared with those in the highest tertile. Participants with decreased TyG (OR, 2.08; 95%CIs, 1.61-2.70) also had increased odds of prediabetes regression compared with those with increased TyG. The cutoff values of initial obesity-related indices were 4.374 for BRI, 0.568 for WHtR, 8.621 for TyG, 1.320 for CI, 0.083 for ABSI, and 106.152 for CVAI, respectively. The AFs were 21.10% for BRI < 4.374, 20.85% for WHtR < 0.568, 17.48% for CVAI < 107.794, and 17.55% for ΔTyG < 0, respectively. Conclusion Low initial BRI, WHtR, and CVAI, as well as TyG reduction, were significantly related to prediabetes regression to normoglycemia, and the AFs were around 20%. Less abdominal fat and insulin resistance reduction would benefit future health outcomes among people with prediabetes.
Collapse
Affiliation(s)
| | - Minjie Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Jiaqi Nie
- School of Public Health, Wuhan University, Wuhan, China
| | - Minzhe Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Gaolei Lu
- School of Public Health, Wuhan University, Wuhan, China
| | - Rui Chen
- School of Public Health, Wuhan University, Wuhan, China
| | - Qiqiang He
- School of Public Health, Wuhan University, Wuhan, China
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Yamazaki H, Streicher SA, Wu L, Fukuhara S, Wagner R, Heni M, Grossman SR, Lenz HJ, Setiawan VW, Marchand LL, Huang BZ. Genetic Evidence Causally Linking Pancreas Fat to Pancreatic Cancer: A Mendelian Randomization Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.20.23288770. [PMID: 37163062 PMCID: PMC10168411 DOI: 10.1101/2023.04.20.23288770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background & Aims Pancreatic ductal adenocarcinoma (PDAC) is highly lethal, and any clues to understanding its elusive etiology could lead to breakthroughs in prevention, early detection, or treatment. Observational studies have shown a relationship between pancreas fat accumulation and PDAC, but the causality of this link is unclear. We therefore investigated whether pancreas fat is causally associated with PDAC using two-sample Mendelian randomization. Methods We leveraged eight genetic variants associated with pancreas fat (P<5×10 -8 ) from a genome-wide association study (GWAS) in the UK Biobank (25,617 individuals), and assessed their association with PDAC in the Pancreatic Cancer Cohort Consortium I-III and the Pancreatic Cancer Case-Control Consortium dataset (8,275 PDAC cases and 6,723 non-cases). Causality was assessed using the inverse-variance weighted method. Although none of these genetic variants were associated with body mass index (BMI) at genome-wide significance, we further conducted a sensitivity analysis excluding genetic variants with a nominal BMI association in GWAS summary statistics from the UK Biobank and the Genetic Investigation of Anthropometric Traits consortium dataset (806,834 individuals). Results Genetically determined higher levels of pancreas fat using the eight genetic variants was associated with increased risk of PDAC. For one standard deviation increase in pancreas fat levels (i.e., 7.9% increase in pancreas fat fraction), the odds ratio of PDAC was 2.46 (95%CI:1.38-4.40, P=0.002). Similar results were obtained after excluding genetic variants nominally linked to BMI (odds ratio:3.79, 95%CI:1.66-8.65, P=0.002). Conclusions This study provides genetic evidence for a causal role of pancreas fat in the pathogenesis of PDAC. Thus, reducing pancreas fat could lower the risk of PDAC.
Collapse
|
24
|
Srinivasan MP, Bhopale KK, Caracheo AA, Kaphalia L, Popov VL, Boor PJ, Kaphalia BS. Dysregulated pancreatic lipid phenotype, inflammation and cellular injury in a chronic ethanol feeding model of hepatic alcohol dehydrogenase-deficient deer mice. Life Sci 2023; 322:121670. [PMID: 37030615 DOI: 10.1016/j.lfs.2023.121670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
AIMS Dysregulation of pancreatic fat and lipotoxic inflammation are common clinical findings in alcoholic chronic pancreatitis (ACP). In this study, we investigated a relationship between dysregulated pancreatic lipid metabolism and the development of injury in a chronic ethanol (EtOH) feeding model of hepatic alcohol dehydrogenase 1- deficient (ADH-) deer mice. METHODS ADH- and hepatic ADH normal (ADH+) deer mice were fed a liquid diet containing 3 % EtOH for three months and received a single gavage of binge EtOH with/without fatty acid ethyl esters (FAEEs) one week before the euthanasia. Plasma and pancreatic tissue were analyzed for lipids including FAEEs, inflammatory markers and adipokines using GC-MS, bioassays/kits, and immunostaining, respectively. Pancreatic morphology and proteins involved in lipogenesis were determined by the H & E staining, electron microscopy and Western blot analysis. KEY FINDINGS Chronic EtOH feeding in ADH- vs. ADH+ deer mice resulted in a significant increase in the levels of pancreatic lipids including FAEEs, adipokines (leptin and resistin), fat infiltration with inflammatory cells and lipid droplet deposition along with the proteins involved in lipogenesis. These changes were exacerbated by an administration of binge EtOH with/without FAEEs in the pancreas of ADH- vs. ADH+ deer mice fed chronic EtOH suggest a metabolic basis for ACP. SIGNIFICANCE These findings suggest that the liver-pancreatic axis plays a crucial role in etiopathogenesis of ACP, as the increased body burden of EtOH due to hepatic ADH deficiency exacerbates pancreatic injury.
Collapse
|
25
|
Mukherjee S, Maheshwari D, Pal R, Sachdeva N. Pancreatic fat in type 2 diabetes: Causal or coincidental? World J Meta-Anal 2023; 11:68-78. [DOI: 10.13105/wjma.v11.i3.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/27/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
|
26
|
Lin D, Wang Z, Li H, Zhang H, Deng L, Ren H, Sun S, Zheng F, Zhou J, Wang M. Automated Measurement of Pancreatic Fat Deposition on Dixon MRI Using nnU-Net. J Magn Reson Imaging 2023; 57:296-307. [PMID: 35635494 DOI: 10.1002/jmri.28275] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Pancreatic fat accumulation may cause or aggravate the process of acute pancreatitis, β-cell dysfunction, T2DM disease, and even be associated with pancreatic tumors. The pathophysiology of fatty pancreas remains overlooked and lacks effective imaging diagnostics. PURPOSE To automatically measure the distribution of pancreatic fat deposition on Dixon MRI in multicenter/population datasets using nnU-Net models. STUDY TYPE Retrospective. POPULATION A total of 176 obese/nonobese subjects (90 males, 86 females; mean age, 27.2 ± 19.7) were enrolled, including a training set (N = 132) and a testing set (N = 44). FIELD STRENGTH/SEQUENCE A 3 T and 1.5 T/gradient echo T1 dual-echo Dixon. ASSESSMENT The segmentation results of four types of nnU-Net models were compared using dice similarity coefficient (DSC), positive predicted value (PPV), and sensitivity. The ground truth was the manual delineation by two radiologists according to in-phase (IP) and opposed-phase (OP) images. STATISTICAL TESTS The group difference of segmentation results of four models were assessed by the Kruskal-Wallis H test with Dunn-Bonferroni comparisons. The interobserver agreement of pancreatic fat fraction measurements across three observers and test-retest reliability of human and machine were assessed by intragroup correlation coefficient (ICC). P < 0.05 was considered statistically significant. RESULTS The three-dimensional (3D) dual-contrast model had significantly improved performance than 2D dual-contrast (DSC/sensitivity) and 3D one-contrast (IP) models (DSC/PPV/sensitivity) and had less errors than 3D one-contrast (OP) model according to higher DSC and PPV (not significant), with a mean DSC of 0.9158, PPV of 0.9105 and sensitivity of 0.9232 in the testing set. The test-retest ICC of this model was above 0.900 in all pancreatic regions, exceeded human. DATA CONCLUSION 3D Dual-contrast nnU-Net aided segmentation of pancreas on Dixon images appears to be adaptable to multicenter/population datasets. It fully automates the assessment of pancreatic fat distribution and has high reliability. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Dingyi Lin
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ziyan Wang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong Li
- School of Medicine, Sir Run Run Shaw Hospital, Department of Endocrinology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongxi Zhang
- School of Medicine, Children's Hospital Binjiang Campus, Department of Radiology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liping Deng
- School of Medicine, Sir Run Run Shaw Hospital, Department of Radiology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong Ren
- School of Medicine, Sir Run Run Shaw Hospital, Department of Radiology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuiya Sun
- School of Medicine, Sir Run Run Shaw Hospital, Department of Endocrinology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fenping Zheng
- School of Medicine, Sir Run Run Shaw Hospital, Department of Endocrinology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiaqiang Zhou
- School of Medicine, Sir Run Run Shaw Hospital, Department of Endocrinology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min Wang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China.,School of Medicine, Sir Run Run Shaw Hospital, Department of Endocrinology, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Ashik T, Lee V, Atanes P, Persaud SJ. Alterations in mouse visceral adipose tissue mRNA expression of islet G-protein-coupled receptor ligands in obesity. Diabet Med 2022; 39:e14978. [PMID: 36245259 PMCID: PMC9828549 DOI: 10.1111/dme.14978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/04/2022] [Accepted: 10/14/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Adipose tissue mass expansion in obesity leads to alterations in expression and secretion of adipokines, some of which may alter islet function by binding to G-protein-coupled receptors (GPCRs) expressed by islets. We have therefore quantified expression of mRNAs encoding islet GPCR ligands in visceral adipose tissue retrieved from lean and diet-induced obese mice to determine alterations in islet GPCR ligand mRNAs in obesity. METHODS Epididymal adipose tissue was retrieved from C57BL/6 mice that had been maintained on a control-fat diet (10% fat) or high-fat diet (60% fat) for 16 weeks and RT-qPCR was used to quantify mRNAs encoding ligands for islet GPCRs. RESULTS Of the 155 genes that encode ligands for islet GPCRs, 45 and 40 were expressed in visceral adipose tissue retrieved from lean and obese mice respectively. The remaining mRNAs were either expressed at trace level (0.0001% to 0.001% relative to Actb expression) or absent (<0.0001%). Obesity was associated with significant alterations in GPCR ligand mRNA expression in visceral adipose tissue, some of which encode for peptides with established effects on islet function (e.g. neuropeptide Y), or for GPCR ligands that have not previously been investigated for their effects on islets (e.g. (C-C motif) ligand 4; Ccl4). CONCLUSION Mouse visceral adipose tissue showed significant alterations in expression of mRNAs encoding islet GPCR ligands in obesity. Our data point to ligands of interest for future research on adipose-islet crosstalk via secreted ligands acting at islet GPCRs. Such research may identify islet GPCRs with therapeutic potential for T2D.
Collapse
Affiliation(s)
- Tanyel Ashik
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Vivian Lee
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Patricio Atanes
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Shanta J. Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| |
Collapse
|
28
|
Leiu KH, Poppitt SD, Miles-Chan JL, Sequeira IR. Fatty Pancreas and Cardiometabolic Risk: Response of Ectopic Fat to Lifestyle and Surgical Interventions. Nutrients 2022; 14:nu14224873. [PMID: 36432559 PMCID: PMC9693202 DOI: 10.3390/nu14224873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Ectopic fat accumulation in non-adipose organs, such as the pancreas and liver, is associated with an increased risk of cardiometabolic disease. While clinical trials have focused on interventions to decrease body weight and liver fat, ameliorating pancreatic fat can be crucial but successful intervention strategies are not yet defined. We identified twenty-two published studies which quantified pancreatic fat during dietary, physical activity, and/or bariatric surgery interventions targeted at body weight and adipose mass loss alongside their subsequent effect on metabolic outcomes. Thirteen studies reported a significant decrease in body weight, utilising weight-loss diets (n = 2), very low-energy diets (VLED) (n = 2), isocaloric diets (n = 1), a combination of diet and physical activity (n = 2), and bariatric surgery (n = 5) including a comparison with VLED (n = 1). Surgical intervention achieved the largest decrease in pancreatic fat (range: -18.2% to -67.2%) vs. a combination of weight-loss diets, isocaloric diets, and/or VLED (range: -10.2% to -42.3%) vs. diet and physical activity combined (range: -0.6% to -3.9%), with a concurrent decrease in metabolic outcomes. While surgical intervention purportedly is the most effective strategy to decrease pancreas fat content and improve cardiometabolic health, the procedure is invasive and may not be accessible to most individuals. Given that dietary intervention is the cornerstone for the prevention of adverse metabolic health, the alternative approaches appear to be the use of weight-loss diets or VLED meal replacements, which are shown to decrease pancreatic fat and associated cardiometabolic risk.
Collapse
Affiliation(s)
- Kok Hong Leiu
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- High Value Nutrition, National Science Challenge, Auckland 1010, New Zealand
| | - Sally D. Poppitt
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- High Value Nutrition, National Science Challenge, Auckland 1010, New Zealand
- Department of Medicine, University of Auckland, Auckland 1010, New Zealand
- Riddet Centre of Research Excellence (CoRE) for Food and Nutrition, Palmerston North 4442, New Zealand
| | - Jennifer L. Miles-Chan
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- High Value Nutrition, National Science Challenge, Auckland 1010, New Zealand
- Riddet Centre of Research Excellence (CoRE) for Food and Nutrition, Palmerston North 4442, New Zealand
| | - Ivana R. Sequeira
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland 1024, New Zealand
- High Value Nutrition, National Science Challenge, Auckland 1010, New Zealand
- Correspondence: ; Tel.: +64-09-6301162
| |
Collapse
|
29
|
Effects of adrenergic-stimulated lipolysis and cytokine production on in vitro mouse adipose tissue-islet interactions. Sci Rep 2022; 12:15831. [PMID: 36138030 PMCID: PMC9499973 DOI: 10.1038/s41598-022-18262-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/08/2022] [Indexed: 11/08/2022] Open
Abstract
Inflammatory cytokines and non-esterified fatty acids (NEFAs) are obesity-linked factors that disturb insulin secretion. The aim of this study was to investigate whether pancreatic adipose tissue (pWAT) is able to generate a NEFA/cytokine overload within the pancreatic environment and as consequence to impact on insulin secretion. Pancreatic fat is a minor fat depot, therefore we used high-fat diet (HFD) feeding to induce pancreatic steatosis in mice. Relative Adipoq and Lep mRNA levels were higher in pWAT of HFD compared to chow diet mice. Regardless of HFD, Adipoq and Lep mRNA levels of pWAT were at least 10-times lower than those of epididymal fat (eWAT). Lipolysis stimulating receptors Adrb3 and Npr1 were expressed in pWAT and eWAT, and HFD reduced their expression in eWAT only. In accordance, HFD impaired lipolysis in eWAT but not in pWAT. Despite expression of Npr mRNA, lipolysis was stimulated solely by the adrenergic agonists, isoproterenol and adrenaline. Short term co-incubation of islets with CD/HFD pWAT did not alter insulin secretion. In the presence of CD/HFD eWAT, glucose stimulated insulin secretion only upon isoproterenol-induced lipolysis, i.e. in the presence of elevated NEFA. Isoproterenol augmented Il1b and Il6 mRNA levels both in pWAT and eWAT. These results suggest that an increased sympathetic activity enhances NEFA and cytokine load of the adipose microenvironment, including that of pancreatic fat, and by doing so it may alter beta-cell function.
Collapse
|
30
|
Yamazaki H, Tauchi S, Machann J, Haueise T, Yamamoto Y, Dohke M, Hanawa N, Kodama Y, Katanuma A, Stefan N, Fritsche A, Birkenfeld AL, Wagner R, Heni M. Fat Distribution Patterns and Future Type 2 Diabetes. Diabetes 2022; 71:1937-1945. [PMID: 35724270 DOI: 10.2337/db22-0315] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022]
Abstract
Fat accumulation in the liver, pancreas, skeletal muscle, and visceral bed relates to type 2 diabetes (T2D). However, the distribution of fat among these compartments is heterogenous and whether specific distribution patterns indicate high T2D risk is unclear. We therefore investigated fat distribution patterns and their link to future T2D. From 2,168 individuals without diabetes who underwent computed tomography in Japan, this case-cohort study included 658 randomly selected individuals and 146 incident cases of T2D over 6 years of follow-up. Using data-driven analysis (k-means) based on fat content in the liver, pancreas, muscle, and visceral bed, we identified four fat distribution clusters: hepatic steatosis, pancreatic steatosis, trunk myosteatosis, and steatopenia. In comparisons with the steatopenia cluster, the adjusted hazard ratios for incident T2D were 4.02 (95% CI 2.27-7.12) for the hepatic steatosis cluster, 3.38 (1.65-6.91) for the pancreatic steatosis cluster, and 1.95 (1.07-3.54) for the trunk myosteatosis cluster. The clusters were replicated in 319 German individuals without diabetes who underwent MRI and metabolic phenotyping. The distribution of the glucose area under the curve across the four clusters found in Germany was similar to the distribution of T2D risk across the four clusters in Japan. Insulin sensitivity and insulin secretion differed across the four clusters. Thus, we identified patterns of fat distribution with different T2D risks presumably due to differences in insulin sensitivity and insulin secretion.
Collapse
Affiliation(s)
- Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinichi Tauchi
- Department of Radiology, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Jürgen Machann
- Section on Experimental Radiology, Department of Radiology, Eberhard-Karls University, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Tobias Haueise
- Section on Experimental Radiology, Department of Radiology, Eberhard-Karls University, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
| | - Yosuke Yamamoto
- Department of Healthcare Epidemiology, School of Public Health, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mitsuru Dohke
- Department of Health Checkup and Promotion, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Nagisa Hanawa
- Department of Health Checkup and Promotion, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Yoshihisa Kodama
- Department of Radiology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Akio Katanuma
- Center for Gastroenterology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Norbert Stefan
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine, Eberhard-Karls University, Tübingen, Germany
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine, Eberhard-Karls University, Tübingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine, Eberhard-Karls University, Tübingen, Germany
| | - Róbert Wagner
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine, Eberhard-Karls University, Tübingen, Germany
- German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- Division of Diabetology, Endocrinology and Nephrology, Department of Internal Medicine, Eberhard-Karls University, Tübingen, Germany
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| |
Collapse
|
31
|
Della Pepa G, Brancato V, Costabile G, Salamone D, Corrado A, Vitale M, Cavaliere C, Mancini M, Salvatore M, Luongo D, Riccardi G, Rivellese AA, Annuzzi G, Bozzetto L. An Isoenergetic Multifactorial Diet Reduces Pancreatic Fat and Increases Postprandial Insulin Response in Patients With Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2022; 45:1935-1942. [PMID: 35862001 DOI: 10.2337/dc22-0605] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/12/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To compare the effect of an isocaloric multifactorial diet with a diet rich in monounsaturated fatty acids (MUFA) and similar macronutrient composition on pancreatic fat (PF) and postprandial insulin response in type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS According to a randomized controlled parallel-group design, 39 individuals with T2D, 35-75 years old, in satisfactory blood glucose control, were assigned to an 8 week isocaloric intervention with a multifactorial diet rich in MUFA, polyunsaturated fatty acids, fiber, polyphenols, and vitamins (n = 18) or a MUFA-rich diet (n = 21). Before/after the intervention, PF content was measured by the proton-density fat fraction using a three-dimensional mDIXON MRI sequence, and plasma insulin and glucose concentrations were measured over a 4 h test meal with a similar composition as the assigned diet. RESULTS After 8 weeks, PF significantly decreased after the multifactorial diet (from 15.7 ± 6.5% to 14.1 ± 6.3%; P = 0.024), while it did not change after the MUFA diet (from 17.1 ± 10.1% to 18.6 ± 10.6%; P = 0.139) with a significant difference between diets (P = 0.014). Postprandial glucose response was similar in the two groups. Early postprandial insulin response (incremental postprandial areas under the curve [iAUC0-120]) significantly increased with the multifactorial diet (from 36,340 ± 34,954 to 44,138 ± 31,878 pmol/L/min; P = 0.037), while it did not change significantly in the MUFA diet (from 31,754 ± 18,446 to 26,976 ± 12,265 pmol/L/min; P = 0.178), with a significant difference between diets (P = 0.023). Changes in PF inversely correlated with changes in early postprandial insulin response (r = -0.383; P = 0.023). CONCLUSIONS In patients with T2D, an isocaloric multifactorial diet, including several beneficial dietary components, markedly reduced PF. This reduction was associated with an improved postprandial insulin response.
Collapse
Affiliation(s)
- Giuseppe Della Pepa
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Giuseppina Costabile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| | - Dominic Salamone
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Alessandra Corrado
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Marilena Vitale
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Marcello Mancini
- Institute of Biostructure and Bioimaging of the National Research Council, Naples, Italy
| | | | - Delia Luongo
- Institute of Biostructure and Bioimaging of the National Research Council, Naples, Italy
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| | - Giovanni Annuzzi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| | - Lutgarda Bozzetto
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| |
Collapse
|
32
|
Horsley V. Adipocyte plasticity in tissue regeneration, repair, and disease. Curr Opin Genet Dev 2022; 76:101968. [PMID: 35988318 DOI: 10.1016/j.gde.2022.101968] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022]
Abstract
Mammalian tissue repair forms a scar that fills the injured area with a fibrotic lesion, limiting tissue function. Adipocytes, lipid-filled cells, well-known for energy storage and endocrine functions, can reside adjacent to or within many tissues, and are emerging as critical regulators of tissue repair. In this review, the plasticity and function of adipocytes to tissue repair and fibrosis in four tissues: skin, heart, skeletal muscle, and mammary gland, will be discussed. The dynamic nature of adipocytes as they release bioactive products, lipids, and adipokines, and their ability to form contractile fibroblasts, is emerging as an essential regulator of wound healing and tumorigenesis in multiple tissues. Thus, modulation of adipocytes may provide therapeutic avenues for regenerative medicine and cancer.
Collapse
Affiliation(s)
- Valerie Horsley
- Department of Molecular and Cell Biology, Yale University, New Haven, CT, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
33
|
Reiterer M, Gilani A, Lo JC. Pancreatic Islets as a Target of Adipokines. Compr Physiol 2022; 12:4039-4065. [PMID: 35950650 DOI: 10.1002/cphy.c210044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Rising rates of obesity are intricately tied to the type 2 diabetes epidemic. The adipose tissues can play a central role in protection against or triggering metabolic diseases through the secretion of adipokines. Many adipokines may improve peripheral insulin sensitivity through a variety of mechanisms, thereby indirectly reducing the strain on beta cells and thus improving their viability and functionality. Such effects will not be the focus of this article. Rather, we will focus on adipocyte-secreted molecules that have a direct effect on pancreatic islets. By their nature, adipokines represent potential druggable targets that can reach the islets and improve beta-cell function or preserve beta cells in the face of metabolic stress. © 2022 American Physiological Society. Compr Physiol 12:1-27, 2022.
Collapse
Affiliation(s)
- Moritz Reiterer
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - James C Lo
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
34
|
Hummel J, Machann J, Dannecker C, Kullmann S, Birkenfeld AL, Häring HU, Peter A, Fritsche A, Wagner R, Heni M. Eight weeks of empagliflozin does not affect pancreatic fat content and insulin secretion in people with prediabetes. Diabetes Obes Metab 2022; 24:1661-1666. [PMID: 35475570 DOI: 10.1111/dom.14733] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Julia Hummel
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Radiology, Section on Experimental Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Corinna Dannecker
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Peter
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Robert Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Diabetes Center (Deutsches Diabetes-Zentrum/DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center, Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| |
Collapse
|
35
|
Linder T, Eppel D, Kotzaeridi G, Rosicky I, Yerlikaya-Schatten G, Kiss H, Weißhaupt K, Henrich W, Bozkurt L, Tura A, Roden M, Göbl CS. Fatty liver indices and their association with glucose metabolism in pregnancy - An observational cohort study. Diabetes Res Clin Pract 2022; 189:109942. [PMID: 35691476 DOI: 10.1016/j.diabres.2022.109942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/15/2022] [Accepted: 06/06/2022] [Indexed: 11/27/2022]
Abstract
AIMS Non-invasive hepatic steatosis indices can be used to assess the risk for metabolic (dysfunction) associated fatty liver disease (MAFLD). This may be helpful to detect metabolic disorders in pregnancy, specifically gestational diabetes (GDM). We aimto examine the association of these indices with parameters of glucose metabolism. METHODS 109 women underwent a metabolic characterization at 16 weeks of gestation andwere classified according to the fatty-liver index (FLI) andhepatic-steatosis index (HSI) into low (G1), intermediate (G2) and high risk (G3). At 26 weeks, participants received an oral glucose tolerance test (OGTT) to assess insulin action, β-cell function and GDM status. RESULTS Both MAFLD indices wereassociated with impaired insulin sensitivityand compensatory increase of insulin release. G3 groups showedimpaired insulin action. The higher circulating insulin concentrations were not able to compensate for insulin resistance in women with higher MAFLD scores, resulting in an increased risk of GDM(OR: 1.05, 95% CI 1.03 to 1.08, p < 0.001 for FLI). MAFLD scores were associated with fetal overgrowth. CONCLUSIONS Maternal MAFLD represents a high-risk obstetric condition. Hepatic steatosis indices are associated with impaired glucose regulation and may provide a useful tool for early risk assessment for impaired glucose metabolism.
Collapse
Affiliation(s)
- Tina Linder
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Daniel Eppel
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Grammata Kotzaeridi
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Ingo Rosicky
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | | | - Herbert Kiss
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Karen Weißhaupt
- Clinic of Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Wolfgang Henrich
- Clinic of Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Latife Bozkurt
- Department of Metabolic Disorders and Nephrology, Hietzing Hospital, Vienna, Austria
| | - Andrea Tura
- Metabolic Unit, CNR Institute of Neuroscience, Padova, Italy
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Christian S Göbl
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria; Clinic of Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
36
|
Differential regulation of mRNAs and lncRNAs related to lipid metabolism in Duolang and Small Tail Han sheep. Sci Rep 2022; 12:11157. [PMID: 35778462 PMCID: PMC9249921 DOI: 10.1038/s41598-022-15318-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
The function of long non-coding RNA (lncRNA) can be achieved through the regulation of target genes, and the deposition of fat is regulated by lncRNA. Fat has an important effect on meat quality. However, there are relatively few studies on lncRNAs in the subcutaneous adipose tissue of Duolang sheep and Small Tail Han sheep. In this study, RNA-Seq technology and bioinformatics methods were used to identify and analyze the lncRNA and mRNA in the subcutaneous adipose tissue of the two breeds of sheep. The results showed that 107 lnRNAs and 1329 mRNAs were differentially expressed. The differentially expressed genes and lncRNA target genes were significantly enriched in the biosynthesis of unsaturated fatty acids signaling pathway, fatty acid metabolism, adipocyte differentiation and other processes related to fat deposition. Among them, LOC105616076, LOC114118103, LOC105607837, LOC101116622, and LOC105603235 target FADS1, SCD, ELOVL6, HSD17B12 and HACD2, respectively. They play a key regulatory role in the biosynthesis of unsaturated fatty acids. This study lays a foundation for the study of the molecular mechanism of lncRNA on fat development, and has reference value for studying the differences in fat deposition between Duolang sheep and Small Tail Han sheep.
Collapse
|
37
|
Wagner R, Eckstein SS, Yamazaki H, Gerst F, Machann J, Jaghutriz BA, Schürmann A, Solimena M, Singer S, Königsrainer A, Birkenfeld AL, Häring HU, Fritsche A, Ullrich S, Heni M. Metabolic implications of pancreatic fat accumulation. Nat Rev Endocrinol 2022; 18:43-54. [PMID: 34671102 DOI: 10.1038/s41574-021-00573-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/15/2022]
Abstract
Fat accumulation outside subcutaneous adipose tissue often has unfavourable effects on systemic metabolism. In addition to non-alcoholic fatty liver disease, which has received considerable attention, pancreatic fat has become an important area of research throughout the past 10 years. While a number of diagnostic approaches are available to quantify pancreatic fat, multi-echo Dixon MRI is currently the most developed method. Initial studies have shown associations between pancreatic fat and the metabolic syndrome, impaired glucose metabolism and type 2 diabetes mellitus. Pancreatic fat is linked to reduced insulin secretion, at least under specific circumstances such as prediabetes, low BMI and increased genetic risk of type 2 diabetes mellitus. This Review summarizes the possible causes and metabolic consequences of pancreatic fat accumulation. In addition, potential therapeutic approaches for addressing pancreatic fat accumulation are discussed.
Collapse
Affiliation(s)
- Robert Wagner
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sabine S Eckstein
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Hajime Yamazaki
- Section of Clinical Epidemiology, Department of Community Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Felicia Gerst
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Benjamin Assad Jaghutriz
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Potsdam-Rehbrücke, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Michele Solimena
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stephan Singer
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral, and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Susanne Ullrich
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Tübingen, Germany.
- Department of Internal Medicine, Division of Diabetology, Endocrinology, and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany.
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
38
|
Zhao YF. Free fatty acid receptors in the endocrine regulation of glucose metabolism: Insight from gastrointestinal-pancreatic-adipose interactions. Front Endocrinol (Lausanne) 2022; 13:956277. [PMID: 36246919 PMCID: PMC9554507 DOI: 10.3389/fendo.2022.956277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Glucose metabolism is primarily controlled by pancreatic hormones, with the coordinated assistance of the hormones from gastrointestine and adipose tissue. Studies have unfolded a sophisticated hormonal gastrointestinal-pancreatic-adipose interaction network, which essentially maintains glucose homeostasis in response to the changes in substrates and nutrients. Free fatty acids (FFAs) are the important substrates that are involved in glucose metabolism. FFAs are able to activate the G-protein coupled membrane receptors including GPR40, GPR120, GPR41 and GPR43, which are specifically expressed in pancreatic islet cells, enteroendocrine cells as well as adipocytes. The activation of FFA receptors regulates the secretion of hormones from pancreas, gastrointestine and adipose tissue to influence glucose metabolism. This review presents the effects of the FFA receptors on glucose metabolism via the hormonal gastrointestinal-pancreatic-adipose interactions and the underlying intracellular mechanisms. Furthermore, the development of therapeutic drugs targeting FFA receptors for the treatment of abnormal glucose metabolism such as type 2 diabetes mellitus is summarized.
Collapse
|
39
|
Filippatos TD, Alexakis K, Mavrikaki V, Mikhailidis DP. Nonalcoholic Fatty Pancreas Disease: Role in Metabolic Syndrome, "Prediabetes," Diabetes and Atherosclerosis. Dig Dis Sci 2022; 67:26-41. [PMID: 33469809 DOI: 10.1007/s10620-021-06824-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Fat accumulation in the pancreas associated with obesity and the metabolic syndrome (MetS) has been defined as "non-alcoholic fatty pancreas disease" (NAFPD). The aim of this review is to describe the association of NAFPD with obesity, MetS, type 2 diabetes mellitus (T2DM) and atherosclerosis and also increase awareness regarding NAFPD. Various methods are used for the detection and quantification of pancreatic fat accumulation that may play a significant role in the differences that have been observed in the prevalence of NAFPD. Endoscopic ultrasound provides detailed images of the pancreas and its use is expected to increase in the future. Obesity and MetS have been recognized as NAFPD risk factors. NAFPD is strongly associated with non-alcoholic fatty liver disease (NAFLD) and it seems that the presence of both may be related with aggravation of NAFLD. A role of NAFPD in the development of "prediabetes" and T2DM has also been suggested by most human studies. Accumulation of fat in pancreatic tissue possibly initiates a vicious cycle of beta-cell deterioration and further pancreatic fat accumulation. Additionally, some evidence indicates a correlation between NAFPD and atherosclerotic markers (e.g., carotid intima-media thickness). Weight loss and bariatric surgery decreases pancreatic triglyceride content but pharmacologic treatments for NAFPD have not been evaluated in specifically designed studies. Hence, NAFPD is a marker of local fat accumulation possibly associated with beta-cell function impairment, carbohydrate metabolism disorders and atherosclerosis.
Collapse
Affiliation(s)
- T D Filippatos
- Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Medicine, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece.
| | - K Alexakis
- Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Medicine, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece
| | - V Mavrikaki
- Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Medicine, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece
| | - D P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, NW3 2QG, UK.,Mohammed Bin Rashid University (MBRU) of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
40
|
Abstract
Pancreatic islets are the body's central rheostat that regulates glucose homeostasis through the production of different hormones, including β cell-derived insulin. During obesity-induced type 2 diabetes (T2D), islet β cells become dysfunctional and inadequate insulin secretion no longer ensures glycemic control. T2D is associated with a chronic low-grade inflammation that manifests in several metabolic organs including the pancreatic islets. Growing evidence suggests that components of the innate immune system, and especially macrophages, play a crucial role in regulating islet homeostasis. Yet, the phenotypes and functions of islet macrophages in physiology and during T2D have only started to attract attention and remain unclear. In this review, the current knowledge about islet inflammation and macrophages will be summarized in humans and rodent models. Recent findings on the cellular and molecular mechanisms involved in islet remodeling and β cell function during obesity and T2D will be discussed.
Collapse
Affiliation(s)
- Joyceline Cuenco
- Centre de Recherche des Cordeliers, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, Paris, France
| | - Elise Dalmas
- Centre de Recherche des Cordeliers, INSERM, IMMEDIAB Laboratory, Sorbonne Université, Université de Paris, Paris, France.
| |
Collapse
|
41
|
Dietz B, Machann J, Agrawal V, Heni M, Schwab P, Dienes J, Reichert S, Birkenfeld AL, Häring HU, Schick F, Stefan N, Fritsche A, Preissl H, Schölkopf B, Bauer S, Wagner R. Detection of diabetes from whole-body MRI using deep learning. JCI Insight 2021; 6:146999. [PMID: 34591793 PMCID: PMC8663560 DOI: 10.1172/jci.insight.146999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/29/2021] [Indexed: 12/21/2022] Open
Abstract
Obesity is one of the main drivers of type 2 diabetes, but it is not uniformly associated with the disease. The location of fat accumulation is critical for metabolic health. Specific patterns of body fat distribution, such as visceral fat, are closely related to insulin resistance. There might be further, hitherto unknown, features of body fat distribution that could additionally contribute to the disease. We used machine learning with dense convolutional neural networks to detect diabetes-related variables from 2371 T1-weighted whole-body MRI data sets. MRI was performed in participants undergoing metabolic screening with oral glucose tolerance tests. Models were trained for sex, age, BMI, insulin sensitivity, HbA1c, and prediabetes or incident diabetes. The results were compared with those of conventional models. The area under the receiver operating characteristic curve was 87% for the type 2 diabetes discrimination and 68% for prediabetes, both superior to conventional models. Mean absolute regression errors were comparable to those of conventional models. Heatmaps showed that lower visceral abdominal regions were critical in diabetes classification. Subphenotyping revealed a group with high future diabetes and microalbuminuria risk.Our results show that diabetes is detectable from whole-body MRI without additional data. Our technique of heatmap visualization identifies plausible anatomical regions and highlights the leading role of fat accumulation in the lower abdomen in diabetes pathogenesis.
Collapse
Affiliation(s)
- Benedikt Dietz
- Department of Computer Science, ETH Zürich, Zürich, Switzerland
| | - Jürgen Machann
- Department of Radiology, Section on Experimental Radiology, Eberhard-Karls University Tübingen, Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Vaibhav Agrawal
- Werner Siemens Imaging Center, Tübingen, Germany.,Max Planck Institute for Intelligent Systems, Department of Empirical Inference, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Patrick Schwab
- Institute of Robotics and Intelligent Systems, ETH Zürich, Zürich, Switzerland
| | - Julia Dienes
- Department of Gynecology and Obstetrics, University Hospital Tübingen, Tübingen, Germany
| | - Steffen Reichert
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Fritz Schick
- Department of Radiology, Section on Experimental Radiology, Eberhard-Karls University Tübingen, Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Bernhard Schölkopf
- Max Planck Institute for Intelligent Systems, Department of Empirical Inference, Tübingen, Germany
| | - Stefan Bauer
- Max Planck Institute for Intelligent Systems, Department of Empirical Inference, Tübingen, Germany.,Department of Intelligent Systems, KTH Stockholm, Stockholm, Sweden
| | - Robert Wagner
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research, Neuherberg, Germany.,Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
42
|
Weaver JR, Odanga JJ, Breathwaite EK, Treadwell ML, Murchinson AC, Walters G, Fuentes DP, Lee JB. An increase in inflammation and islet dysfunction is a feature of prediabetes. Diabetes Metab Res Rev 2021; 37:e3405. [PMID: 33463010 DOI: 10.1002/dmrr.3405] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/26/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022]
Abstract
AIMS Type 2 diabetes (T2D) is a global health problem that will be diagnosed in almost 300 million people by 2025 according to the World Health Organization. Before being diagnosed with T2D, individuals may have glucose levels above normal but below the diabetic range. This condition is known as prediabetes. Studies showed that people with prediabetes had an increase in several pro-inflammatory cytokines in their serum and in their fasting glucose levels. The answer remains unclear when inflammation begins in the pancreas and islets, and what is the extent of this inflammation. METHODS Subjects with haemoglobin A1c levels from 5.7% to 6.4% were classified as pre-diabetic. Sections of pancreas and isolated islets from normal donors and donors with prediabetes were tested for markers of inflammation and glucose-stimulated insulin secretion (GSIS). RESULTS Gene and protein expression of the inflammatory markers resistin, interleukin-1 beta, tumour necrosis factor-alpha, interleukin-6, and monocyte chemoattractant protein-1 increased in donors with prediabetes compared to normal donors. GSIS response was significantly decreased in pre-diabetic islets compared to normal islets. Donors with prediabetes also had decreased expression of CD163+ cells but not CD68+ cells. CONCLUSIONS Based on our findings, inflammation and islet dysfunction may be more significant than originally thought in people with prediabetes. Rather than being in a normal state before diabetes occurs, it appears that subjects are already in an early diabetic condition resembling more closely T2D.
Collapse
Affiliation(s)
- Jessica R Weaver
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Justin J Odanga
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Erick K Breathwaite
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Michelle L Treadwell
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Angela C Murchinson
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Gary Walters
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Danette P Fuentes
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| | - Jung Bok Lee
- Institute of Regenerative Medicine, LifeNet Health, Virginia Beach, Virginia, USA
| |
Collapse
|
43
|
Streicher SA, Lim U, Park SL, Li Y, Sheng X, Hom V, Xia L, Pooler L, Shepherd J, Loo LWM, Darst BF, Highland HM, Polfus LM, Bogumil D, Ernst T, Buchthal S, Franke AA, Setiawan VW, Tiirikainen M, Wilkens LR, Haiman CA, Stram DO, Cheng I, Le Marchand L. Genome-wide association study of pancreatic fat: The Multiethnic Cohort Adiposity Phenotype Study. PLoS One 2021; 16:e0249615. [PMID: 34329319 PMCID: PMC8323875 DOI: 10.1371/journal.pone.0249615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/15/2021] [Indexed: 01/26/2023] Open
Abstract
Several studies have found associations between higher pancreatic fat content and adverse health outcomes, such as diabetes and the metabolic syndrome, but investigations into the genetic contributions to pancreatic fat are limited. This genome-wide association study, comprised of 804 participants with MRI-assessed pancreatic fat measurements, was conducted in the ethnically diverse Multiethnic Cohort-Adiposity Phenotype Study (MEC-APS). Two genetic variants reaching genome-wide significance, rs73449607 on chromosome 13q21.2 (Beta = -0.67, P = 4.50x10-8) and rs7996760 on chromosome 6q14 (Beta = -0.90, P = 4.91x10-8) were associated with percent pancreatic fat on the log scale. Rs73449607 was most common in the African American population (13%) and rs79967607 was most common in the European American population (6%). Rs73449607 was also associated with lower risk of type 2 diabetes (OR = 0.95, 95% CI = 0.89-1.00, P = 0.047) in the Population Architecture Genomics and Epidemiology (PAGE) Study and the DIAbetes Genetics Replication and Meta-analysis (DIAGRAM), which included substantial numbers of non-European ancestry participants (53,102 cases and 193,679 controls). Rs73449607 is located in an intergenic region between GSX1 and PLUTO, and rs79967607 is in intron 1 of EPM2A. PLUTO, a lncRNA, regulates transcription of an adjacent gene, PDX1, that controls beta-cell function in the mature pancreas, and EPM2A encodes the protein laforin, which plays a critical role in regulating glycogen production. If validated, these variants may suggest a genetic component for pancreatic fat and a common etiologic link between pancreatic fat and type 2 diabetes.
Collapse
Affiliation(s)
- Samantha A. Streicher
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Unhee Lim
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - S. Lani Park
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Yuqing Li
- Department of Epidemiology and Biostatistics, University of California – San Francisco, San Francisco, California, United States of America
| | - Xin Sheng
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Victor Hom
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Lucy Xia
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Loreall Pooler
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - John Shepherd
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Lenora W. M. Loo
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Burcu F. Darst
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Heather M. Highland
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Linda M. Polfus
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - David Bogumil
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Thomas Ernst
- University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Steven Buchthal
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Adrian A. Franke
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Veronica Wendy Setiawan
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Maarit Tiirikainen
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Lynne R. Wilkens
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Daniel O. Stram
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, University of California – San Francisco, San Francisco, California, United States of America
| | - Loïc Le Marchand
- University of Hawaii Cancer Center, University of Hawaii at Mānoa, Honolulu, Hawaii, United States of America
| |
Collapse
|
44
|
Zhang K, Tao C, Xu J, Ruan J, Xia J, Zhu W, Xin L, Ye H, Xie N, Xia B, Li C, Wu T, Wang Y, Schroyen M, Xiao X, Fan J, Yang S. CD8 + T Cells Involved in Metabolic Inflammation in Visceral Adipose Tissue and Liver of Transgenic Pigs. Front Immunol 2021; 12:690069. [PMID: 34322121 PMCID: PMC8311854 DOI: 10.3389/fimmu.2021.690069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Anti-inflammatory therapies have the potential to become an effective treatment for obesity-related diseases. However, the huge gap of immune system between human and rodent leads to limitations of drug discovery. This work aims at constructing a transgenic pig model with higher risk of metabolic diseases and outlining the immune responses at the early stage of metaflammation by transcriptomic strategy. We used CRISPR/Cas9 techniques to targeted knock-in three humanized disease risk genes, GIPRdn , hIAPP and PNPLA3I148M . Transgenic effect increased the risk of metabolic disorders. Triple-transgenic pigs with short-term diet intervention showed early symptoms of type 2 diabetes, including glucose intolerance, pancreatic lipid infiltration, islet hypertrophy, hepatic lobular inflammation and adipose tissue inflammation. Molecular pathways related to CD8+ T cell function were significantly activated in the liver and visceral adipose samples from triple-transgenic pigs, including antigen processing and presentation, T-cell receptor signaling, co-stimulation, cytotoxicity, and cytokine and chemokine secretion. The similar pro-inflammatory signaling in liver and visceral adipose tissue indicated that there might be a potential immune crosstalk between the two tissues. Moreover, genes that functionally related to liver antioxidant activity, mitochondrial function and extracellular matrix showed distinct expression between the two groups, indicating metabolic stress in transgenic pigs' liver samples. We confirmed that triple-transgenic pigs had high coincidence with human metabolic diseases, especially in the scope of inflammatory signaling at early stage metaflammation. Taken together, this study provides a valuable large animal model for the clinical study of metaflammation and metabolic diseases.
Collapse
Affiliation(s)
- Kaiyi Zhang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Cong Tao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianping Xu
- The Ministry of Health Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinxue Ruan
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, China
| | - Jihan Xia
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wenjuan Zhu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Leilei Xin
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huaqiong Ye
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ning Xie
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Boce Xia
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chenxiao Li
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tianwen Wu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanfang Wang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | | | - Xinhua Xiao
- The Ministry of Health Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiangao Fan
- Shanghai Key Laboratory of Children's Digestion and Nutrition, Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shulin Yang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Key Laboratory of Animal Genetics Breeding and Reproduction, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
45
|
Obesity-induced changes in human islet G protein-coupled receptor expression: Implications for metabolic regulation. Pharmacol Ther 2021; 228:107928. [PMID: 34174278 DOI: 10.1016/j.pharmthera.2021.107928] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022]
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that are the targets for many different classes of pharmacotherapy. The islets of Langerhans are central to appropriate glucose homeostasis through their secretion of insulin, and islet function can be modified by ligands acting at the large number of GPCRs that islets express. The human islet GPCRome is not a static entity, but one that is altered under pathophysiological conditions and, in this review, we have compared expression of GPCR mRNAs in human islets obtained from normal weight range donors, and those with a weight range classified as obese. We have also considered the likely outcomes on islet function that the altered GPCR expression status confers and the possible impact that adipokines, secreted from expanded fat depots, could have at those GPCRs showing altered expression in obesity.
Collapse
|
46
|
Ulasoglu C, Tekin ZN, Akan K, Yavuz A. Does Nonalcoholic Pancreatic Steatosis Always Correlate with Nonalcoholic Fatty Liver Disease? Clin Exp Gastroenterol 2021; 14:269-275. [PMID: 34149286 PMCID: PMC8205613 DOI: 10.2147/ceg.s317340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To identify the correlation of nonalcoholic pancreatic steatosis (NAPS) with nonalcoholic fatty liver disease (NAFLD) in an outpatient group. Based on its metabolic and imaging properties, NAPS has been increasingly recognized in recent years; however, its interaction with NAFLD is still not clear. PATIENTS AND METHODS In this cross-sectional observational study, 345 consecutive patients without any chronic illness who were referred to the senior radiologist for abdominal ultrasound (US) were included. The US report showed hepatic and pancreatic echogenicity. The patients' demographic, anthropometric, and laboratory data were collected from medical records. RESULTS Overall, NAPS and NAFLD were seen in 227 (65.8%) and 219 (63.5%) patients, respectively. Normal echogenicity was noted in 74 (21.4%) patients. Forty-four patients (12.8%) had steatotic liver without NAPS, 52 (15.1%) had steatotic pancreas without NAFLD, and 175 (50.7%) had steatosis in both organs. The discordance in steatosis grading between NAPS and NAFLD was 55.1%. Insulin resistance was present in 8.7, 26.7, 19, and 61.3% of patients with no steatosis, only NAFLD, only NAPS, and steatosis in both organs, respectively. Evident NAFLD and NAPS having grade 2 and 3 steatosis were present in 15.3% and 29.0% of the study group, respectively. Cholecystolithiasis was present in 6.8, 13.6, and 28.8% of patients with normal echogenic pancreas, only NAFLD, and only NAPS, respectively (p=0.01). CONCLUSION Based on the ultrasonographic, clinical, demographic, and anthropometric features of the included patients, we found that NAPS did not fully accompany nonalcoholic fatty liver. Despite severe pancreatic steatosis, more than a quarter of cases had normal liver echogenicity. Insulin resistance frequency was insignificantly higher in NAFLD than NAPS (p=0.694). The significantly higher frequency of cholecystolithiasis in NAPS needs further large-scale studies. The inconsistency of steatosis degree in NAPS and NAFLD in >50% cases may reflect differences in the pathophysiology of these two clinical entities.
Collapse
Affiliation(s)
- Celal Ulasoglu
- Department of Gastroenterology, Medeniyet University, Goztepe Education and Research Hospital, Istanbul, Turkey
| | - Zeynep Nilufer Tekin
- Department of Radiology, Medeniyet University, Goztepe Education and Research Hospital, Istanbul, Turkey
| | - Kubra Akan
- Department of Gastroenterology, Medeniyet University, Goztepe Education and Research Hospital, Istanbul, Turkey
| | - Arda Yavuz
- Department of Gastroenterology, Medeniyet University, Goztepe Education and Research Hospital, Istanbul, Turkey
| |
Collapse
|
47
|
Gezginci-Oktayoglu S, Sancar S, Karatug-Kacar A, Bolkent S. miR-375 induces adipogenesis through targeting Erk1 in pancreatic duct cells under the influence of sodium palmitate. J Cell Physiol 2021; 236:3881-3895. [PMID: 33107061 DOI: 10.1002/jcp.30129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
The goal of this study was to research long-term saturated fatty acid overexposure that can induce differentiation of pancreatic duct cells into adipocytes and also into β-cells. The important findings can be summarized as follows: (i) adipogenesis and early stage β-cell differentiation were stimulated in duct cells under lipotoxicity and glucolipotoxicity conditions, (ii) miR-375 expression was upregulated while its target Erk1 was downregulated and miR-375 inhibitor upregulated Erk1 while expression of adipogenesis markers was downregulated in duct cells under both conditions, (iii) apoptosis was induced in β and duct cells under both conditions, (iv) lipotoxicity induced proliferation of co-cultured β-cells. These findings suggest that long-term saturated fatty acid overexposure may cause intrapancreatic fat accumulation by inducing differentiation of duct cells into adipocytes and it may contributes to β-cell compensation by stimulating the early stage of β-cell differentiation in duct cells. In addition, miR-375 may have the potential to be a new target in the treatment of Type 2 diabetes, and NAFPD due to its role in the adipogenesis of duct cells.
Collapse
Affiliation(s)
- Selda Gezginci-Oktayoglu
- Biology Department, Molecular Biology Section, Faculty of Science, Istanbul University, Vezneciler, Istanbul, Turkey
| | - Serap Sancar
- Biology Department, Molecular Biology Section, Faculty of Science, Istanbul University, Vezneciler, Istanbul, Turkey
| | - Ayse Karatug-Kacar
- Biology Department, Molecular Biology Section, Faculty of Science, Istanbul University, Vezneciler, Istanbul, Turkey
| | - Sehnaz Bolkent
- Biology Department, Molecular Biology Section, Faculty of Science, Istanbul University, Vezneciler, Istanbul, Turkey
| |
Collapse
|
48
|
Rehman SU, Schallschmidt T, Rasche A, Knebel B, Stermann T, Altenhofen D, Herwig R, Schürmann A, Chadt A, Al-Hasani H. Alternative exon splicing and differential expression in pancreatic islets reveals candidate genes and pathways implicated in early diabetes development. Mamm Genome 2021; 32:153-172. [PMID: 33880624 PMCID: PMC8128753 DOI: 10.1007/s00335-021-09869-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/03/2021] [Indexed: 12/29/2022]
Abstract
Type 2 diabetes (T2D) has a strong genetic component. Most of the gene variants driving the pathogenesis of T2D seem to target pancreatic β-cell function. To identify novel gene variants acting at early stage of the disease, we analyzed whole transcriptome data to identify differential expression (DE) and alternative exon splicing (AS) transcripts in pancreatic islets collected from two metabolically diverse mouse strains at 6 weeks of age after three weeks of high-fat-diet intervention. Our analysis revealed 1218 DE and 436 AS genes in islets from NZO/Hl vs C3HeB/FeJ. Whereas some of the revealed genes present well-established markers for β-cell failure, such as Cd36 or Aldh1a3, we identified numerous DE/AS genes that have not been described in context with β-cell function before. The gene Lgals2, previously associated with human T2D development, was DE as well as AS and localizes in a quantitative trait locus (QTL) for blood glucose on Chr.15 that we reported recently in our N2(NZOxC3H) population. In addition, pathway enrichment analysis of DE and AS genes showed an overlap of only half of the revealed pathways, indicating that DE and AS in large parts influence different pathways in T2D development. PPARG and adipogenesis pathways, two well-established metabolic pathways, were overrepresented for both DE and AS genes, probably as an adaptive mechanism to cope for increased cellular stress. Our results provide guidance for the identification of novel T2D candidate genes and demonstrate the presence of numerous AS transcripts possibly involved in islet function and maintenance of glucose homeostasis.
Collapse
Affiliation(s)
- Sayeed Ur Rehman
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Tanja Schallschmidt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Axel Rasche
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Birgit Knebel
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Torben Stermann
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Delsi Altenhofen
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Ralf Herwig
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,German Institute of Human Nutrition, Potsdam, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany. .,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
49
|
Bouazizi K, Zarai M, Dietenbeck T, Aron-Wisnewsky J, Clément K, Redheuil A, Kachenoura N. Abdominal adipose tissue components quantification in MRI as a relevant biomarker of metabolic profile. Magn Reson Imaging 2021; 80:14-20. [PMID: 33872732 DOI: 10.1016/j.mri.2021.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/15/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Abnormal accumulation of adipose tissue (AT) alters the metabolic profile and underlies cardiovascular complications. Conventional measures provide global measurements for the entire body. The purpose of this study was to propose a new approach to quantify the amount and type of truncal AT automatically from MRI in metabolic patients and controls. MATERIALS AND METHODS DIXON acquisitions were performed at 1.5 T in 30 metabolic syndrome (MS) (59 ± 6 years), 12 obese (50 ± 11 years), 35 type 2 diabetes (T2DM) patients (56 ± 11 years) and 19 controls (52 ± 11 years). AT was segmented into: subcutaneous AT "SAT", visceral AT "VAT", deep VAT "dVAT", peri-organ VAT "pVAT" using active contours and k-means clustering algorithms. Subsequently, organ AT infiltration index "oVAT" was calculated as the normalized fat signal magnitude in organs. RESULTS Excellent intra- and inter-operator reproducibility was obtained for AT segmentation. MS and obese patients had the highest amount of total AT. SAT increased in MS (1144 ± 621 g) and T2DM patients (1024 ± 634 g), and twice the level of SAT in controls (505 ± 238 g), and further increased in obese patients (1429 ± 621 g). While VAT, pVAT and dVAT increased to a similar degree in the metabolic patients compared to controls, the oVAT index was able to differentiate controls from MS and T2DM patients and to discriminate the three metabolic patient groups (p < 0.01). Local AT sub-types were not related to BMI in all groups except for SAT in controls (p = 0.03). CONCLUSION Reproducible truncal AT sub-types quantification using 3D MRI was able to characterize patients with metabolic diseases. It may serve in the future as a non-invasive predictor of cardiovascular complications in such patients.
Collapse
Affiliation(s)
- Khaoula Bouazizi
- Institute of Cardiometabolism And Nutrition (ICAN), La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne University, INSERM 1146, CNRS 7371, Laboratoire d'Imagerie Biomédicale, Paris, France.
| | - Mohamed Zarai
- Institute of Cardiometabolism And Nutrition (ICAN), La Pitié-Salpêtrière Hospital, Paris, France
| | - Thomas Dietenbeck
- Institute of Cardiometabolism And Nutrition (ICAN), La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne University, INSERM 1146, CNRS 7371, Laboratoire d'Imagerie Biomédicale, Paris, France
| | - Judith Aron-Wisnewsky
- Institute of Cardiometabolism And Nutrition (ICAN), La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Université, INSERM, Nutrition and Obesities; approches systémiques (NutriOmique), Pitié-Salpêtrière Hospital, Nutrition Department, Paris, France
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesities; approches systémiques (NutriOmique), Pitié-Salpêtrière Hospital, Nutrition Department, Paris, France
| | - Alban Redheuil
- Institute of Cardiometabolism And Nutrition (ICAN), La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne University, INSERM 1146, CNRS 7371, Laboratoire d'Imagerie Biomédicale, Paris, France; Unité d'Imagerie Cardiovasculaire et Thoracique (ICT), Pitié-Salpêtrière Hospital, Paris, France
| | - Nadjia Kachenoura
- Institute of Cardiometabolism And Nutrition (ICAN), La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne University, INSERM 1146, CNRS 7371, Laboratoire d'Imagerie Biomédicale, Paris, France
| |
Collapse
|
50
|
Barroso Oquendo M, Siegel-Axel D, Gerst F, Lorza-Gil E, Moller A, Wagner R, Machann J, Fend F, Königsrainer A, Heni M, Häring HU, Ullrich S, Birkenfeld AL. Pancreatic fat cells of humans with type 2 diabetes display reduced adipogenic and lipolytic activity. Am J Physiol Cell Physiol 2021; 320:C1000-C1012. [PMID: 33788629 DOI: 10.1152/ajpcell.00595.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity, especially visceral fat accumulation, increases the risk of type 2 diabetes (T2D). The purpose of this study was to investigate the impact of T2D on the pancreatic fat depot. Pancreatic fat pads from 17 partial pancreatectomized patients (PPP) were collected, pancreatic preadipocytes isolated, and in vitro differentiated. Patients were grouped using HbA1c into normal glucose tolerant (NGT), prediabetic (PD), and T2D. Transcriptome profiles of preadipocytes and adipocytes were assessed by RNAseq. Insulin sensitivity was estimated by quantifying AKT phosphorylation on Western blots. Lipogenic capacity was assessed with oil red O staining, lipolytic activity via fatty acid release. Secreted factors were measured using ELISA. Comparative transcriptome analysis of preadipocytes and adipocytes indicates defective upregulation of genes governing adipogenesis (NR1H3), lipogenesis (FASN, SCD, ELOVL6, and FADS1), and lipolysis (LIPE) during differentiation of cells from T2D-PPP. In addition, the ratio of leptin/adiponectin mRNA was higher in T2D than in NGT-PPP. Preadipocytes and adipocytes of NGT-PPP were more insulin sensitive than T2D-PPP cells in regard to AKT phosphorylation. Triglyceride accumulation was similar in NGT and T2D adipocytes. Despite a high expression of the receptors NPR1 and NPR2 in NGT and T2D adipocytes, lipolysis was stimulated by ANP 1.74-fold in NGT cells only. This stimulation was further increased by the PDE5 inhibitor dipyridamole (3.09-fold). Dipyridamole and forskolin increased lipolysis receptor independently 1.88-fold and 1.48-fold, respectively, solely in NGT cells. In conclusion, the metabolic status persistently affects differentiation and lipolysis of pancreatic adipocytes. These alterations could aggravate the development of T2D.
Collapse
Affiliation(s)
- Morgana Barroso Oquendo
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Dorothea Siegel-Axel
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Felicia Gerst
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Estela Lorza-Gil
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Anja Moller
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Robert Wagner
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany.,Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Susanne Ullrich
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases, Helmholtz Center Munich, Eberhard-Karls-University of Tübingen, Neuherberg, Germany.,Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
| |
Collapse
|